Next Article in Journal
Expression and Effects of High-Mobility Group Box 1 in Cervical Cancer
Next Article in Special Issue
Adipose-Derived Stem Cells: A Review of Signaling Networks Governing Cell Fate and Regenerative Potential in the Context of Craniofacial and Long Bone Skeletal Repair
Previous Article in Journal
Geminal Brønsted Acid Ionic Liquids as Catalysts for the Mannich Reaction in Water
Previous Article in Special Issue
Lyophilized Platelet-Rich Fibrin (PRF) Promotes Craniofacial Bone Regeneration through Runx2
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Signaling Pathways in Cartilage Repair

1
Laboratory of Immunorheumatology and Tissue Regeneration/RAMSES, Rizzoli Orthopaedic Institute, via di Barbiano 1/10, Bologna 40136, Italy
2
Department of Medical and Surgical Sciences, University of Bologna, Bologna 40138, Italy
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2014, 15(5), 8667-8698; https://doi.org/10.3390/ijms15058667
Submission received: 25 February 2014 / Revised: 28 April 2014 / Accepted: 4 May 2014 / Published: 15 May 2014
(This article belongs to the Special Issue Signal Transduction of Tissue Repair)

Abstract

:
In adult healthy cartilage, chondrocytes are in a quiescent phase characterized by a fine balance between anabolic and catabolic activities. In ageing, degenerative joint diseases and traumatic injuries of cartilage, a loss of homeostatic conditions and an up-regulation of catabolic pathways occur. Since cartilage differentiation and maintenance of homeostasis are finely tuned by a complex network of signaling molecules and biophysical factors, shedding light on these mechanisms appears to be extremely relevant for both the identification of pathogenic key factors, as specific therapeutic targets, and the development of biological approaches for cartilage regeneration. This review will focus on the main signaling pathways that can activate cellular and molecular processes, regulating the functional behavior of cartilage in both physiological and pathological conditions. These networks may be relevant in the crosstalk among joint compartments and increased knowledge in this field may lead to the development of more effective strategies for inducing cartilage repair.

1. Introduction

Articular cartilage is an avascular, highly specialized tissue constituted by an extensive extracellular matrix (ECM) mainly composed of two types of macromolecules, collagen (types II, IX and XI) and proteoglycan (aggrecan) [1]. The biophysical properties of cartilage derive from this highly organized fibrillar framework, that supplies shape, strength, tensile stiffness and compressive resistance to the tissue [2]. The synthesis, maintenance and degradation of ECM proteins are coordinated by chondrocytes, the only resident cell type in cartilage [2].
In healthy adult cartilage, chondrocytes are in a quiescent phase characterized by a fine balance between synthetic and catabolic activities [1]. In ageing, degenerative joint diseases and traumatic injuries of cartilage, a loss of homeostatic conditions and an up-regulation of catabolic pathways occur [1,3].
In osteoarthritis (OA), the most prevalent chronic joint disease, several different factors are recognized as main mediators and/or effectors of progressive cartilage loss, including proinflammatory cytokines (IL-1, TNF) [46], chemokines, [711], extracellular matrix degrading enzymes, such as metalloproteinases (MMPs) and aggrecanases (A Disintegrin And Metalloproteinase with Thrombospondin Motifs-ADAMTS), which act as downstream key players in the inflammatory signal cascade.
A further hallmark of degenerated cartilage is the modification of chondrocyte differentiation stage. Indeed, chondrocyte phenotype switches toward a hypertrophic phenotype [1,12], thus recapitulating some of the physiological differentiation steps occurring in growth plates and endochondral ossification.
Hypertrophic chondrocytes are characterized by the expression of terminal differentiation markers, including Runt-related transcription factor 2 (RUNX-2), Collagen X, MMP-13 and Indian hedgehog (Ihh) [1318] and by the acquisition of an “autolytic” phenotype marked by their ability to induce the degradation of pericellular cartilage matrix [13,19]. Conversely, the expression of hypertrophy-related genes is downregulated by cartilage protective mechanism such as Dickkopf (Dkk-1), which acts by inhibiting Wnt signaling [20].
The fate of chondrocytes to remain within cartilage or undergo hypertrophic maturation prior to ossification is also subject to complex regulation by the interplay of the FGF, TGF-β, BMP and Wnt signaling pathways. In particular, whereas FGF signaling accelerates the rate of terminal hypertrophic differentiation, BMPs have been shown to hinder this process. Therefore FGF signals act as antagonists of BMP signaling and negatively regulate Ihh expression [21].
Given the well-known limited ability of cartilage to self-regenerate, damage induced by aging, injury and pathological conditions undergoes unsuccessful reparative processes, resulting in a nonfunctional newly formed tissue [22,23]. Due to this lack of self-regeneration, several strategies have been applied to improve the treatment of cartilage lesions: surgical techniques, transplantation approaches, tissue engineering and bioregeneration technologies [24].
Since cartilage differentiation and maintenance of homeostatic conditions are finely tuned by a complex network of signaling molecules and biophysical factors, shedding light on these mechanisms appears to be extremely relevant for both the identification of key pathogenic factors as specific therapeutic targets and the development of biological approaches for improving cartilage regeneration.
This review focuses on the crucial pathways that drive cartilage physiopathology. In particular, we will discuss the critical signals that can activate cellular and molecular processes in both cartilage regeneration and degradation: transforming growth factor-β (TGF-β), bone morphogenetic proteins (BMPs), fibroblast growth factors (FGFs), hypoxia-related factors (HIF), Wnt/β-catenin, nuclear factor kappa B (NF-κB), mitogen-activated protein kinase (MAPK) and hedgehog (Hh) cascades.

2. Transforming Growth Factor-β (TGF-β), Bone Morphogenetic Protein (BMP), Insulin Growth Factor (IGF) and Fibroblast Growth Factor (FGF) Pathways

The development, growth, maintenance and repair of articular cartilage are controlled by several very stringent signaling pathways, triggered by numerous bioactive growth factors. They are needed to properly regulate chondrogenesis and a combination of them, working synergistically, is probably necessary for the cartilage reparative procedure, enhancing of cartilage matrix synthesis as in the case of BMP-7 and IGF-I [25], and differential regulation of their own and each other’s gene expression and protein production in vitro as for IGF-I, FGF-2, and TGF-β [26]. Among these molecules those of the TGF-β family play a prominent role (reviewed by [27]).
The TGF-β superfamily is comprised of more than forty members, also including the BMPs [28]. It is noteworthy that TGF-β1 is one of the main molecules considered to be anabolic for cartilage [2931], together with Insulin Growth Factor (IGF)-1 [32], Fibroblast Growth Factor (FGF)-2 [33] and BMP-7 [34]. Conversely, TGF-β has been shown to be involved in cartilage degeneration during ageing and OA. These conflicting actions depend on the alternative activation of different signaling pathways [13,19,3537].
TGF-β signals via its type II receptor which then engages the type I receptors. These receptors are called activin-like kinase (ALK)1 and ALK5 [35,3840] and respectively they activate the Smad 1-5-8 pathway and phosphorylate Smad 2–3 [35,39].
Strong evidence suggests that these two activation pathways are master regulators of chondrocyte phenotypic change and differentiation progression [35]. This hypothesis is based mainly on animal studies but it is corroborated by confirmation studies on human OA tissues [29,30,41,42].
ALK5 activation by TGF-β engagement and subsequent signaling via Smad 2–3 contributes to the maintenance of the stable quiescent phase of chondrocytes and the induction of aggrecan and collagen II production. Smad 2 and 3 exert an inhibitory effect on chondrocyte hypertrophy [30,43], which represents the phenotypic hallmark of terminal differentiated chondrocytes. A similar phenotypic modification occurs in OA and also in ageing chondrocytes [13,44,45] and it has been shown to be associated with a reduced expression of ALK5 leading to a break of the chondrocyte quiescent state and the induction of the terminal differentiation of chondrocytes [13].
Conversely, the activation of the Smad 1-5-8 pathway by ALK1 cooperates with RUNX-2 to stimulate hypertrophic differentiation with the consequent production of Collagen X, MMP13, osteopontin, alkaline phosphatase, osteocalcin and vascular endothelial growth factor (VEGF) by chondrocytes [42,46].
Recent elegant studies by the group at Radboud University have demonstrated a shift in the ALK1/ALK5 ratio occurring in ageing and during OA both in humans and in mice [35]. In ageing and in OA, a loss of the TGF-β receptor ALK5 reduced the phosphorylation of Smad 2–3, whereas only a small decrease in ALK1 expression is documented [35], therefore a relative predominance of Smad 1-5-8 signaling is operating in ageing and OA cartilage, thus promoting the hypertrophic differentiation.
The above-mentioned studies underline the complexity of the various actions of TGF-β in cartilage homeostasis and OA development. A great deal of data from mouse studies can be applied to human pathology with caution. Nonetheless, during OA development (and in ageing) chondrocytes are under the simultaneous influence of various stimuli that probably induce reciprocal opposing effects, the net sum of which determines the final metabolic response.
BMPs are involved in all phases of chondrogenesis and are essential for the endochondral bone formation (reviewed by [47]). These activities are carried out by regulating Smad 1-5-8 and Smad 4 which are critical transcription regulators [46]. Several BMPs (namely BMP-2, -4–6, -11) have been detected in normal and OA cartilage [48]. Although BMPs are recognized as protective factors, being able to play an important role in regeneration of cartilage, they have been shown to also be involved in chondrocyte hypertrophy and matrix degradation. Indeed, BMP-2 promotes chondrocyte proliferation and matrix synthesis [4952] and controls chondrogenesis through the regulation of the expression and activity of SRY-related high-mobility-group box transcription factor (SOX) 9 [5355]. The role of BMP-2 in enhancing cartilage repair and counteracting cartilage damage is also underlined by studies in animals. In a mouse model of IL-1-induced cartilage injury, BMP-2 increased the collagen II and aggrecan expression. Moreover, blocking BMP activity resulted in a reduced synthesis of proteoglycan [52] and increased cartilage damage [56]. Conversely, BMP-2 induces hypertrophic differentiation of chondrocytes and may promote cartilage degradation by elevating MMP-13 expression, as observed in OA cartilage [57,58]. The requirement of BMPs for chondrocyte terminal differentiation is highlighted by the evidence that loss of Smad 1 and 5 or inhibition of the Smad 1/5/8 signaling cascade blocks the differentiation of chondrocytes and leads to severe cartilage defects [59,60].
Among the BMPs synthesized by human chondrocytes, BMP-7 has been shown to have both anabolic and anti-catabolic effects on cartilage, being able to induce extracellular matrix synthesis by chondrocytes and counteract the catabolic effect induced on these cells by IL-1, IL-6 and fibronectin fragments [6165]. Evidence concerning the critical role of cell-matrix interaction in the regulation of BMP signaling has been reported (reviewed by [66,67]). In cartilage, hyaluronan-dependent pericellular matrix is critical for BMP-7 mediated response [68,69] and a very recent study demonstrated that the expression of CD44, the principal chondrocyte hyaluronan receptor, supports the cellular response to lower concentrations of BMP-7 [69].
Recently, differential actions of BMP-2 and BMP-7 on the outcome of chondrocyte differentiation have been reported: BMP-2 stimulated chondrocyte hypertrophy, whereas BMP-7 prevented chondrocyte hypertrophy and maintained chondrogenic potential [58]. BMP-7 is already marketed as an agent able to speed up bone healing after fracture [70]. Overall these results provide evidence that currently, BMPs can be considered key players in cartilage repair and promising candidates for clinical application in the field of cartilage and bone regenerative medicine [71,72].
The role of IGF-I in articular cartilage metabolism has been extensively investigated in both physiological and pathological conditions [7375]. When added exogenously to monolayers or explant cultures of normal articular cartilage from a variety of species, IGF-I induces a plethora of anabolic effects and decreases catabolic responses [76,77]. In particular, IGF-I has been reported to predominantly stimulate matrix synthesis with a minor effect on mitotic activity in articular chondrocytes [78,79] and to accelerate proliferation and differentiation of cartilage progenitor cells in cultures of neonatal mandibular condyles [80]. Furthermore, in the presence of IGF-I, the chondrocyte phenotype was maintained and cells did not progress to the hypertrophic stage [81].
The need for IGF-I to maintain articular cartilage integrity is supported by an in vivo study in rats in which chronic IGF-I deficiency led to the development of articular cartilage lesions [82]. In animal models, it was shown that IGF-I enhanced the repair of extensive cartilage defects and protected the synovial membrane from chronic inflammation [83,84]. The interactions between IGF-1 and TGF-β1 on the proliferation and differentiation of chondrocytes have been reported by several studies [8587], which also highlighted the ability of TGF-β to increase the number of IGF-I receptors in chondrocytes without changing their affinity [86].
Fukumoto and coworkers showed that IGF-1 in combination with TGF-β1 enhanced total cartilage production compared with IGF-1 alone. In particular, TGF-β has been found to act early to induce chondrogenesis, whereas IGF-I enhanced and maintained proliferation, increasing total cartilage formation [77]. Similarly, MSC chondrogenic differentiation is induced by IGF-I but is enhanced when IGF-I and TGF-β1 are used in combination [88,89].
However, the ability of chondrocytes to respond to IGF-I decreases with age [9092] and in OA [91,93,94]. Evidence suggests an uncoupling of IGF-I responsiveness in OA, indicating that, in OA cartilage organ cultures, IGF-I is able to robustly stimulate proteoglycan synthesis at saturating doses, but it is unable to modulate proteoglycan catabolism [95]. Despite the diminished ability of IGF-I to decrease catabolism in aged and OA cartilage, a combination of IGF-I and BMP-7 results in greater repair potential by enhancing matrix synthesis than either growth factor alone [25,96].
Fibroblast growth factors (FGFs) constitute another family of relevant growth factors for cartilage development and homeostasis [97]. In particular, two members of this family, FGF-2 and FGF-18, have been implicated in the regulation of cartilage remodeling [97,98]. Conflicting results have been obtained concerning the effect of FGF-2 on chondrocytes. Different studies have shown a regenerative effect on cartilage defect treated with FGF-2, in animal models [99102]. These findings might result from the strong mitogenic effect exerted on chondrocytes by this growth factor [103], which, on the other hand, appears to be ineffective at inducing a successful regeneration of cartilage extracellular matrix [103]. In agreement with anti-anabolic/catabolic effects of FGF-2, studies performed on human chondrocytes [104107] showed the ability of FGF-2 to up-regulate MMP-13, ADAMTS-4, -5, inhibit cartilage matrix production, antagonize activity of anabolic factors (e.g., BMP-7) and stimulate proinflammatory cytokines, such as IL-1 and TNF.
In contrast to FGF-2, the anabolic effect of FGF-18 in cartilage has been well established [108111]. In porcine and human cartilage, FGF-18 has been shown to act as an inducer of cell proliferation and extracellular matrix synthesis [108]. Furthermore, in a rat OA model, intra-articular injections of FGF-18 increased cartilage formation and reduced cartilage degeneration scores [111].

3. Hypoxia-Inducible Factor (HIF) Pathway

Healthy articular cartilage is typically an avascular tissue. Chondrocyte survival in this hypoxic condition requires the activation of adaptive strategies that promote and sustain tissue function in a low oxygen environment. Hypoxic response is mainly mediated by hypoxia inducible factor (HIF) family members (HIF-1α, -2α, -3α). The first member of this transcription factor family to be identified was HIF-1α [112].
In the presence of normal oxygen levels, HIF-1α is hydroxilated and degraded, whereas, under hypoxic conditions, hydroxilation is inhibited and HIF-1α is free to form a heterodimer with the constitutive HIF-1β unit. This complex binds specific target gene consensus sequences and promotes their transcription [113]. In cartilage, HIF-1α has been shown to be essential for chondrocyte growth arrest and survival [114] and appears to be involved in the regulation of angiogenetic factor expression, mainly VEGF [114116], thus modulating an essential step in endochondral bone formation. Furthermore, HIF-1α contributes to the maintenance of ECM homeostasis, inducing the gene expression of two main matrix components: collagen II and aggrecan [117,118].
The crucial role of HIF-1α in controlling the survival of hypoxic cartilage has been highlighted by further experimental evidence, underlining the central importance of HIF-1 in supporting the chondrocyte energy generation, via increased glucose uptake and by regulating the activity of glycolytic enzymes [119121], the synthesis of cartilage matrix [117] and the activation of a protective mechanism enabling the prevention of chondrocyte cell death induced by IL-1β [121].
In damaged cartilage, HIF-1α expression has been reported in several studies [120124]. An increasing transcription of HIF-1α in OA cartilage compared to normal samples was shown, particularly in the late-stage of the disease. Consistent with this evidence, subsequent studies reported a growing number of HIF-1α-positive chondrocytes during OA progression [120] and a higher expression of HIF-1α mRNA in degenerated cartilage compared to uninjured cartilage [121].
In addition to hypoxic conditions, HIF-1α expression can be up-regulated by other factors, including inflammatory cytokines (IL-1 and TNF-β), reactive oxygen species and mechanical loading [121,125131], which are all recognized as key players in cartilage damage.
Since HIF-1α has a pivotal role in supporting chondrocyte survival and cartilage homeostasis, these findings have led researchers to consider HIF-1α as a stress-inducible responder, rather than merely as a hypoxia-inducible factor, which potentially acts in order to maintain the chondroprotective functions challenged by the detrimental conditions occurring in the OA joint environment. Overall, these findings characterize HIF-1α as a key factor for chondrocyte survival promoting compensatory mechanisms in response to catabolic modifications of OA cartilage.
Another member of the hypoxic inducible factor family, HIF-2α, has subsequently been recognized. HIF-2α and HIF-1α have extensive structural homology and both of them are regulated by similar mechanisms [132]. Despite these similarities, these transcriptional factors have been shown to have specific and distinct functions under physiological and pathological conditions [133135].
In healthy cartilage, hypoxic conditions appear to promote the up-regulation of cartilage matrix genes via HIF-2α-mediated SOX-9 induction [136,137].
The distinct, but complementary roles of HIF-1α and HIF-2α have been reported in a study performed on healthy human cartilage: HIF-2α mediates anabolic responses by directly binding a specific SOX-9 regulatory site, HIF-1α induces anti-catabolic effects by modulating the expression of cartilage degrading enzymes and its inhibitors [138]. Furthermore, HIF-2α has been identified as an extensive regulator of endochondral bone formation [139]. This process is marked by sequential steps, including chondrocyte hypertrophic differentiation (characterized by secretion of collagen X), cartilage degradation (via proteinases, mainly MMP-13) and vascular invasion depending on angiogenic stimuli, such as VEGF [140,141]. HIF-2α appears to be a central regulator of all these steps, directly regulating collagen X, MMP-13 and VEGF expression. In addition, further possible transcription targets of HIF-2α related to endochondral ossification have been identified, namely RUNX-2 and Indian hedgehog proteins [139,142].
Conversely, HIF-2α appears to be highly expressed in degenerated cartilage and strongly implicated in catabolic mechanisms leading to cartilage breakdown and endochondral bone formation, which represents a fundamental pathway leading to osteophyte formation, considered as one of the typical OA outcomes [143]. The role of HIF-2α as catabolic inducer of OA cartilage destruction has been clearly demonstrated in studies by Yang et al. [144]. They showed that the adenoviral overexpression of HIF-2α causes progressive cartilage damage directly by up-regulating the expression of various degradative enzymes, including MMP1, MMP3, MMP12, MMP13, ADAMTS4.
The relevance of HIF-2α in OA has been strengthened by additional evidence from human knee joint samples, showing increased expression related to the early and progressive stage of OA development, whereas this factor appears to decrease in the late stage of the disease.
HIFs are also implicated in regulating chondrocyte autophagy. Autophagy is a primary process that degrades and removes damaged and dysfunctional intracellular organelles and molecules, thus protecting cells during stress responses [145147]. Disorders of this mechanism have been found to contribute to the development of aging-related diseases [148,149], indeed this process appears to be compromised in OA cartilage. The role of HIF-1α and HIF-2α in controlling chondrocyte autophagy has been shown by studies demonstrating the ability of HIF-2α to counterbalance the ability of HIF-1α to accelerate chondrocyte autophagy functions [150].
Even if the conflicting role of HIF-2 needs to be clarified, the balance between HIF-1α/HIF-2α activities clearly contributes to the control of cartilage homeostasis.

4. Wnt/β-Catenin Pathway

Wnts (Wingless-type) constitute a large family of 19 secreted glycoproteins involved in the development, growth and homeostasis of different tissues and organs, including joints, bone and cartilage (reviewed in [151154]). Wnts bind receptors of the Frizzled (FZD) family receptors on the plasma membrane to initiate several distinct cascades classified as either canonical or non-canonical, depending on whether β-catenin is involved.
The Wnt pathway, often referred to as the canonical pathway, is the best described and signals through the β-catenin nuclear effector. It is activated upon the binding of Wnt to Frizzled (FZD) receptors and low density lipoprotein receptor-related protein (LRP) 5/6 co-receptors at the cell surface [155,156]. In the absence of appropriate Wnt ligands, β-catenin is phosphorylated using glycogen synthase kinase 3 (GSK3) and, subsequently, targeted for ubiquitination and degradation via the proteasome [157159]. However, in the presence of a Wnt ligand, its binding to the receptor complex initiates the activation of an intracellular protein termed Dishevelled (Dvl) which in turn inhibits GSK3 activity [160]. Therefore, β-catenin is not targeted for degradation; it accumulates and translocates to the nucleus and modulates the transcription of target genes.
The Wnt/β-catenin pathway is tightly regulated by several natural extracellular antagonists of Wnt signaling, namely secreted frizzled related proteins (sFRPs), Wnt inhibitory factors (Wifs), Dickkopf (Dkk) factors and sclerostin (SCL). The inhibition of the signaling cascade essentially occurs through two different mechanisms: the direct binding of sFRPs and Wifs to Wnts, that interferes with the FZD receptor interactions and blocking of the canonical pathway performed through Dkk and SCL, binding to the LRP5/6 Wnt coreceptor [158,159].
Chondrogenesis is highly dependent upon cell–ECM and cell–cell adhesion interactions and it has been shown that Wnt signaling is necessary either for the maintenance of mature articular cartilage phenotype, which is characterized by extended cell survival and absence of differentiation towards hypertrophy (schematically reported in Table 1) or for the hypertrophic maturation in the process of endochondral ossification (schematically reported in Table 2) (reviewed by [151,152,154]).
Wnt signaling has recently been shown to play an additional role in crosstalk between cartilage and subchondral bone. During late embryonic and postnatal development, the rate of trabecular bone formation is strongly dependent on the levels of β-catenin expressed by chondrocytes in the lower hypertrophic zone. Wnts have been extensively recognized as key regulators of bone cartilage and joint development and homeostasis [158,161164].
β-catenin in hypertrophic chondrocytes is necessary for regulating the expression of RANKL to control osteoclast activity in subchondral growth plates. Thus, it may be expected that, during the progression of OA, the observed altered activity of Wnt signaling in chondrocytes may affect the osteoclastic activity in subchondral bone growth plates leading to sclerosis or osteophyte formation at the edges of joints. The alteration of Wnt signaling pathway in chondrocytes appears to modulate key regulatory factors for remodeling of subchondral bone, resulting in its aberrant behavior as observed in the case of OA. Similarly, altered Wnt signaling in subchondral bone may modulate chondrogenic factors that are necessary for maintaining cartilage homeostasis [162].
This evidence has prompted researchers to investigate the potential association between Wnt modifications and OA. Enhanced activation of canonical Wnt pathways in OA human cartilage and in injured cartilage [165167] has been reported. These findings have been confirmed by further studies performed on animal models that report a relationship between β-catenin signaling activation and OA-like phenotype [168170].
Genetic studies have identified some components of the Wnt cascade as candidate genes associated with OA. A single nucleotide polymorphism in the sFRP3 gene, a Wnt antagonist, appears to be linked to an increased risk for hip OA [171], whereas an association between a single LRP5 gene polymorphism and spine OA has been reported [172].
The relevance of Dkk-1 as a key regulator of bone remodeling has been shown. In fact, the blockage of Dkk-1, abrogating Dkk-1-mediated Wnt suppression, reverts from the bone-destructive pattern in a mouse model of RA and induces bone formation and osteophyte growth, thus resembling the bone-forming pattern of OA [173].
Wnt signaling antagonists are also considered as potential biomarkers of OA progression. Elevated Dkk-1 serum levels have been shown to be associated with reduced progression of hip OA in a Caucasian population [174]. Similarly, in patients with knee OA, an inverse correlation between Dkk-1 levels in plasma and synovial fluid and the radiographic severity has been reported [175]. In addition, high serum concentrations of sFRP-3 showed a tendency to be associated with a lower risk of developing hip OA [174].
Extensive studies on Wnt signaling related to bone and cartilage physiology and pathology have undoubtedly led to a body of evidence consistent with a critical role in OA pathogenesis. Nevertheless, considering the complexity of the downstream effects of Wnt and its multiple interplays with several other pathways, further research is needed to better characterize the specific role and relevance of single Wnt agents/antagonists in bone, cartilage and the osteochondral junction.

5. Nuclear Factor-Kappa B (NF-κB) Pathway

The NF-κB transcription factors are a family of ubiquitously expressed molecules that regulate a wide range of immune responses, cellular growth, differentiation and survival both in normal and neoplastic conditions (Reviewed by [176180]). Concerning cartilage, activated NF-κB regulates the expression of several matrix degrading enzymes, thus influencing the amount and remodeling of ECM proteins, and shows indirect positive effects on downstream regulators of terminal chondrocyte differentiation (including β-catenin and RUNX-2).
The transcriptional control exerted by NF-κB, is performed by means of five proteins combined in homo or hetero-dimers (RelA/p65, RelB, c-Rel, NF-κB1/p50 (or p105 as a precursor inactive form of p50) and NF-κB2/p52 (or p100 as a precursor inactive form of p52), whose shared N-terminal domain with Rel-homology is crucial for different activities, such as the interaction with specific inhibitory proteins, the dimerization in active complexes and the translocation to nucleus [177,181].
The inactive form of the NF-κB dimers is located in the cell cytoplasm bound to specific inhibitory (I-κB) molecules and translocates into the nucleus following cell activation. The stimulation induces the phosphorylation of I-κB molecules mediated by I-κB kinases (IKKB) that, following ubiquitination, are degraded by a proteasome complex [177].
Two distinct pathways regulate the activation of the NF-κB signaling cascades: the canonical/classical way (mediated by TNF, Toll-like and T-cell receptors) and the alternative/non-canonical one (mediated by B-cell activating factor-BAFF, CD40 and lymphotoxin-b receptors) [180,182184]. The canonical pathway is crucial for the activation of the innate and inflammatory responses as well as for the inhibition of apoptosis, whereas the alternative pathway is involved in lymphoid organogenesis, B-cell activation and humoral immunity. Once in the nucleus, released NF-κB dimers bind to the promoter region of various genes, including immunomodulatory molecules, cytokines, cyclooxygenase-2 (COX-2), inducible NO-synthase (iNOS) and proteases (MMP), thereby activating their transcription [182,185,186].
The activation of NF-κB signaling in OA chondrocytes is induced by fibronectin fragments, pro-inflammatory mediators (TNF-α or IL-1β), mechanical stress and ageing factors through cytokine and Toll-like membrane receptors [187]. The NF-κB pathway alone or cooperating with other pathways (such as Wnt or BMPs) induces the secretion of various degradative enzymes including various matrix metallo-proteinases [177] and ADAMTS4 and ADAMTS5 [181], leading to articular cartilage destruction [1].
MMP in OA chondrocytes is also induced by various catabolic soluble factors such as IL-1beta, IL-6, IL-8, TNF-α and receptor activator of NF-κB (RANK) ligand (RANKL). These cytokines and chemokines decrease also the synthesis of proteoglycan and collagen and contribute to amplifying NF-κB activation. [6]. NF-κB molecules contribute to the articular changes by inducing type X collagen (involved in chondrocyte hypertrophy) and prostaglandin E2 (PGE2), nitric oxide (NO), nitric oxide synthase (NOS) and cyclooxygenase 2 (COX2), which promote the synthesis of catabolic factors, cartilage inflammation and apoptosis of OA chondrocytes [177,188190].
The protracted activation of NF-κB influences other regulatory transcription factors, such as HIF-2α, RUNX-2 and E74-like factor 3 (ELF3) [191,192]. ELF3 acts as a central mediator of the inflammatory responses triggered by IL-1, TNFα and LPS and modulates the IL-6 expression induced by LPS in vivo [193]. In chondrocytes, ELF3 accounts for the IL-1-mediated repression of the type II collagen [194] and participates in the IL-1-induced MMP-13 expression [195].
The above transcription factors stimulate the production of MMP13 and ADAMTS5 thus favoring the acquisition of chondrocyte hypertrophic phenotype (characterized by chondrocyte calcification and osteophyte formation) [191]. In these conditions, the NF-κB proteins induce the synthesis of BMP2, IL-8, GRO-α and growth arrest and DNA damage-inducible protein 45beta (GADD45beta) effector molecules [196,197] and, together with ELF3 and HIF-2α pathways, induce the expression of type X collagen, MMP9, MMP13, alkaline phosphatase, VEGF and osteocalcin [198].
Besides its role in cartilage destruction, NF-κB regulates many genes including cytokines, chemokines and adhesion molecules that participate in the pathophysiology of synovial inflammation (synovitis) and bone degradation in OA joints [187].
In synovial cells the activation of NF-κB [199] mediated by different stimuli (cytokines, cartilaginous matrix fragments, stress) [200,201] induces the synthesis of various cytokines and chemokines (IL-1β, IL-6, TNF-α, RANKL, IL-8, RANTES), angiogenic factors (VEGF, FGF-b) and degradative enzymes (MMPs1-13, ADAMTS4, ADAMTS5) that lead to further cartilage destruction and increased synovitis [202].
Concerning bone, IL-1, IL-6, and TNF (produced by both chondrocyte and synovial cells) induce RANKL synthesis and the binding between the RANKL osteoclastogenic cytokine and RANK surface receptor activates NF-κB [203] to produce IL-1, IL-6, and PGE2 and then bone resorption [204].
In normal conditions RANK/RANKL interaction is regulated by the OPG decoy receptor that by blocking RANKL promotes bone formation [205], whereas in OA a decreased OPG/RANKL ratio favors osteoclast activity and the resulting bone resorption [206209].

6. Mitogen-Activated Protein Kinase (MAPK) Pathway

The MAPK pathway is known to control the conversion of a vast number of extracellular stimuli into specific cellular responses, such as embryogenesis, cell proliferation, differentiation, and survival (reviewed by [153,210213]).
Three major classes of MAPKs have been described in mammals: the extracellular signal-regulated kinases (ERK-1/2), the c-jun N-terminal kinase (JNK) and p38 [214]. ERKs are activated by mitogens and growth factors, whereas JNK and p38 are activated by inflammatory cytokines (IL-1 and TNF-α), cellular stress (heat and osmotic), reactive oxygen species, and ultraviolet irradiation [215217].
Activation of ERK, JNK and p38 MAPK signaling cascades coordinates phosphorylation events that result in the activation of transcription factors such as AP-1 (cFos/cJun), ETS, RUNX-2, HIF-2α, and C/EBPβ, which together with NF-κB, regulate the expression of genes involved in catabolic and inflammatory events [218221].
Several studies have shown the requirement of MAPK signaling pathways, during various phases of cartilage formation and maturation and endochondral ossification [210,222,223].
These pathways have been found to play a distinct role in cartilage matrix synthesis and homeostasis and alterations in these signaling pathways (p38 and ERK-1/2, in particular) are reported to play a prominent role in chondrocyte dysfunction as a part of OA pathogenesis and disease progression [213,224]. The activities of p38 are primarily governed by extensive cross-talk with ERK-1/2, resulting in a reciprocal bidirectional equilibrium between ERK-1/2 and p38 phosphorylation [213].
During normal chondrogenesis, the process of cartilage nodule formation is activated by p38s and repressed by ERK-1/2 [225]. In agreement, a strong activation of the ERK-1/2 pathway was observed in the hypertrophic zone of the growth plate, whereas the inhibition of ERK-1/2 delayed hypertrophic differentiation in growth plate chondrocytes during endochondral ossification [210,213]. Furthermore such cross-talk (activation of ERK-1/2 and deactivation of p38) appears to play a role in the induction of hypertrophic changes observed in normal articular chondrocytes in vitro, when co-cultured with subchondral osteoblasts form OA patients or exposed to their conditioned medium [213].
Early and late differentiation processes of osteoblasts are promoted by both p38 and ERK-1/2, whereas all three kinases appear to promote osteoclast differentiation by regulating activator protein 1 (AP-1) as a critical mediator of osteoclastogenesis [223]. Furthermore, ERK1/2 is possibly a central regulator to the effects of pro-inflammatory cytokines IL-1β, IL-6, IL-12, IL-23, and TNF-α that cause joint destruction [226,227].
In vitro, IL-1 leads to the activation of ERK, p38 and JNK and is associated with the up-regulated expression of MMPs in chondrocytes and conversely, ERK1/2 inhibition decreases the release of MMP3 and MMP13 by human chondrocytes stimulated with IL-1β [228]. In addition, IL-1β exhibits inhibitory effects on collagen type II and aggrecan whose synthesis is necessary for cartilage repair. Thus, IL-1β disrupts the balance between catabolic and anabolic activities, which may result in articular cartilage destruction [228], MMP13 is capable of cleaving type II collagen, whereas MMP3 is active against other components of the extracellular matrix, such as fibronectin and laminin.
The MAPK/ERK pathway is also involved in the IL-1β-induced decrease in type II collagen and aggrecan expression in engineered cartilage, thus compromising the effective host tissue integration [229]. The altered expression of MMP3, MMP13, type II collagen and aggrecan induced by IL-1β stimulation in human chondrocytes was significantly and similarly reverted by ERK1 or ERK2 knockdown; no isoform was significantly upregulated after the single knockdown of the other kinase, and the combined knockdown displayed synergistic effects [228].

7. Hedgehog (Hh)/Smoothened (Smo) Pathway

Another signaling pathway strongly involved in both chondrogenesis and chondrocyte proliferation and differentiation in the growth plate during development [230235] is the Hedgehog (Hh) pathway. The ligands of this pathway (Indian Hh (Ihh) and Sonic Hh (Shh)) are expressed by chondrocytes in response to mechanical stress (Ihh is a mechanoresponsive gene) [236]. These ligands engage Patched 1 receptor (Ptch1), terminating, in this way, the inhibitory action of Ptch1 on another cell surface receptor called Smoothened (Smo) [237]. The Hh ligand binding to Ptch1 causes Smo localization and accumulation in the chondrocyte cilia [238] and induces the activation of downstream transcription factors called Gli proteins [239], the final result being the expression of RUNX-2 (a master regulator of chondrocyte hypertrophic differentiation) and, indirectly, the expression of the aggrecanase ADAMTS5 [240]. In chondrocytes, Ihh signaling induces the expression of parathyroid hormone-related peptide (PTHrP) ligand [234] and the engagement of the corresponding parathyroid hormone 1 receptor (PTH1R) results in the repression of both Ihh expression and chondrocyte hypertrophy. Thus, the Ihh/PTHrP feedback loop controls the proliferative zone of the growth plate and regulates the rate of hypertrophy [234,235,241].
Similarly to the growth plate, in mature cartilage the Ihh–PTHrP axis regulates the differentiation program of chondrocytes, by coordinating chondrocyte maturation and hypertrophy [242] and participating in maintenance of the physiological functionality of articular cartilage.
Since OA chondrocytes typically recapitulate some differentiation steps occurring in growth plates, the involvement of hedgehog signaling has been addressed by several studies.
Hh signaling appears to be aberrantly activated in OA cartilage, thus promoting the hypertrophic phenotype of chondrocytes and up-regulation of MMP-13, one of the extracellular matrix-degrading enzymes that play a key role as mediators of cartilage damage in OA [243]. The inhibition of Hh signaling reduces disease severity in OA animal models, thus identifying a potential therapeutic target for OA treatment [240].
The Ihh/PTHrP axis modulation has also been explored as a molecular system for promoting cartilage repair. Ihh gene transfer induces chondrogenic differentiation of mesenchymal stem cells (MSC) [244] and the ability of Shh to induce redifferentiation of dedifferentiated chondrocytes by the stimulation of SOX-9, BMP-2, and IGF expression has been recently reported [245].
Furthermore, the presence of PTHrP in cultured MSC is able to suppress the hypertrophy during “in vitro” chondrogenesis [246]. These findings underline the potential relevance of this pathway to improve outcomes of cartilage regenerative approaches and tissue engineering therapies.

8. Signaling Cross-Talk and Conclusions

The balance between anabolic and catabolic activities in cartilage appears to be driven by many pathways. Over the years, each of these pathways had been extensively investigated and partly clarified, but little is still understood about their interplay.
Nevertheless, it is becoming clear that there is significant cross-talk between the different pathways and that the overall effects on chondrocyte function depend on the balance in activity of multiple signaling proteins.
The expression of Wnt/β-catenin negatively regulates NF-κB; on the contrary, canonical Wnt signaling drives TGF-β/BMP—signaling towards the Alk1, Smad 1-5-8 route. Wnt/β-catenin also interacts with Ihh signaling and all these pathways converge on the activation of RUNX-2 and downstream target gene expression (MMP13, MMP3, and Collagen X), thus inducing the hypertrophic chondrocyte phenotype (Figure 1).
In its turn, Wnt/β-catenin is negatively modulated by HIF-1α and positively modulated by MAPK and HIF-2α and by TGF-β/BMP—signaling via the ALK5, Smad 2–3 pathway and, depending on the prevailing cross regulation of the different routes, a good balance (homeostasis/stable chondrocyte) or a deleterious scenario (damage/chondrocyte hyperthropy) may result (Figure 1) [247250].
The substantial progress made in identifying the pathways regulating anabolic/catabolic response of chondrocytes in developing and adult cartilage has unraveled an ever increasing complexity of the molecular networks involved. The continuous progression of our knowledge regarding the molecular pathways controlling the functional behavior of cartilage, in both physiological and pathological conditions, may lead to the development of more effective strategies for inducing cartilage repair and for the treatment of cartilage-related diseases.

Acknowledgments

This work was partially supported by grants from Rizzoli Orthopaedic Institute (Ricerca Corrente); University of Bologna (RFO); Italian Ministry of Health (Project RF-2009-1498841); PRRU (Emilia-Romagna Region/University of Bologna Project) 2010–2012; “Cinque per mille” Funds and was performed within the framework of ADIPOA EU FP7 (project No. 241719).
The authors gratefully acknowledge Keith Smith for English revision.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Goldring, M.B.; Marcu, K.B. Cartilage homeostasis in health and rheumatic diseases. Arthritis Res. Ther 2009, 11. [Google Scholar] [CrossRef]
  2. Poole, A.R.; Kojima, T.; Yasuda, T.; Mwale, F.; Kobayashi, M.; Laverty, S. Composition and structure of articular cartilage: A template for tissue repair. Clin. Orthop. Relat. Res 2001, 391, S26–S33. [Google Scholar]
  3. Wang, M.; Shen, J.; Jin, H.; Im, H.J.; Sandy, J.; Chen, D. Recent progress in understanding molecular mechanisms of cartilage degeneration during osteoarthritis. Ann. N. Y. Acad. Sci 2011, 1240, 61–69. [Google Scholar]
  4. Goldring, M.B. Osteoarthritis and cartilage: The role of cytokines. Curr. Rheumatol. Rep 2000, 2, 459–465. [Google Scholar]
  5. Melchiorri, C.; Meliconi, R.; Frizziero, L.; Silvestri, T.; Pulsatelli, L.; Mazzetti, I.; Borzi, R.M.; Uguccioni, M.; Facchini, A. Enhanced and coordinated in vivo expression of inflammatory cytokines and nitric oxide synthase by chondrocytes from patients with osteoarthritis. Arthritis Rheum 1998, 41, 2165–2174. [Google Scholar]
  6. Kapoor, M.; Martel-Pelletier, J.; Lajeunesse, D.; Pelletier, J.P.; Fahmi, H. Role of proinflammatory cytokines in the pathophysiology of osteoarthritis. Nat. Rev. Rheumatol 2011, 7, 33–42. [Google Scholar]
  7. Borzi, R.M.; Mazzetti, I.; Marcu, K.B.; Facchini, A. Chemokines in cartilage degradation. Clin. Orthop. Relat. Res 2004, S53–S61. [Google Scholar]
  8. Wei, L.; Kanbe, K.; Lee, M.; Wei, X.; Pei, M.; Sun, X.; Terek, R.; Chen, Q. Stimulation of chondrocyte hypertrophy by chemokine stromal cell-derived factor 1 in the chondro-osseous junction during endochondral bone formation. Dev. Biol 2010, 341, 236–245. [Google Scholar]
  9. Rasheed, Z.; Akhtar, N.; Haqqi, T.M. Advanced glycation end products induce the expression of interleukin-6 and interleukin-8 by receptor for advanced glycation end product-mediated activation of mitogen-activated protein kinases and nuclear factor-kappaB in human osteoarthritis chondrocytes. Rheumatology 2011, 50, 838–851. [Google Scholar]
  10. Wenke, A.K.; Niebler, S.; Grassel, S.; Bosserhoff, A.K. The transcription factor AP-2varepsilon regulates CXCL1 during cartilage development and in osteoarthritis. Osteoarthr. Cartil 2011, 19, 206–212. [Google Scholar]
  11. Chao, P.Z.; Hsieh, M.S.; Cheng, C.W.; Lin, Y.F.; Chen, C.H. Regulation of MMP-3 expression and secretion by the chemokine eotaxin-1 in human chondrocytes. J. Biomed. Sci 2011, 18. [Google Scholar] [CrossRef]
  12. Fosang, A.J.; Beier, F. Emerging Frontiers in cartilage and chondrocyte biology. Best Pract. Res. Clin. Rheumatol 2011, 25, 751–766. [Google Scholar]
  13. Van der Kraan, P.M.; van den Berg, W.B. Chondrocyte hypertrophy and osteoarthritis: Role in initiation and progression of cartilage degeneration? Osteoarthr. Cartil 2012, 20, 223–232. [Google Scholar]
  14. Fuerst, M.; Bertrand, J.; Lammers, L.; Dreier, R.; Echtermeyer, F.; Nitschke, Y.; Rutsch, F.; Schafer, F.K.; Niggemeyer, O.; Steinhagen, J.; et al. Calcification of articular cartilage in human osteoarthritis. Arthritis Rheum 2009, 60, 2694–2703. [Google Scholar]
  15. Little, C.B.; Barai, A.; Burkhardt, D.; Smith, S.M.; Fosang, A.J.; Werb, Z.; Shah, M.; Thompson, E.W. Matrix metalloproteinase 13-deficient mice are resistant to osteoarthritic cartilage erosion but not chondrocyte hypertrophy or osteophyte development. Arthritis Rheum 2009, 60, 3723–3733. [Google Scholar]
  16. Von der Mark, K.; Kirsch, T.; Nerlich, A.; Kuss, A.; Weseloh, G.; Gluckert, K.; Stoss, H. Type X collagen synthesis in human osteoarthritic cartilage. Indication of chondrocyte hypertrophy. Arthritis Rheum 1992, 35, 806–811. [Google Scholar]
  17. Gelse, K.; Ekici, A.B.; Cipa, F.; Swoboda, B.; Carl, H.D.; Olk, A.; Hennig, F.F.; Klinger, P. Molecular differentiation between osteophytic and articular cartilage—Clues for a transient and permanent chondrocyte phenotype. Osteoarthr. Cartil 2012, 20, 162–171. [Google Scholar]
  18. Mueller, M.B.; Tuan, R.S. Functional characterization of hypertrophy in chondrogenesis of human mesenchymal stem cells. Arthritis Rheum 2008, 58, 1377–1388. [Google Scholar]
  19. Van der Kraan, P.M.; Blaney Davidson, E.N.; van den Berg, W.B. A role for age-related changes in TGFβ signaling in aberrant chondrocyte differentiation and osteoarthritis. Arthritis Res. Ther 2010, 12, 201. [Google Scholar]
  20. Leijten, J.C.; Emons, J.; Sticht, C.; van Gool, S.; Decker, E.; Uitterlinden, A.; Rappold, G.; Hofman, A.; Rivadeneira, F.; Scherjon, S.; et al. Gremlin 1, frizzled-related protein, and Dkk-1 are key regulators of human articular cartilage homeostasis. Arthritis Rheum 2012, 64, 3302–3312. [Google Scholar]
  21. Minina, E.; Kreschel, C.; Naski, M.C.; Ornitz, D.M.; Vortkamp, A. Interaction of FGF, Ihh/Pthlh, and BMP signaling integrates chondrocyte proliferation and hypertrophic differentiation. Dev. Cell 2002, 3, 439–449. [Google Scholar]
  22. Caplan, A.I.; Elyaderani, M.; Mochizuki, Y.; Wakitani, S.; Goldberg, V.M. Principles of cartilage repair and regeneration. Clin. Orthop. Relat. Res 1997, 342, 254–269. [Google Scholar]
  23. Hunziker, E.B. Articular cartilage repair: Basic science and clinical progress A review of the current status and prospects. Osteoarthr. Cartil 2002, 10, 432–463. [Google Scholar]
  24. Johnstone, B.; Alini, M.; Cucchiarini, M.; Dodge, G.R.; Eglin, D.; Guilak, F.; Madry, H.; Mata, A.; Mauck, R.L.; Semino, C.E.; et al. Tissue engineering for articular cartilage repair—The state of the art. Eur. Cell Mater 2013, 25, 248–267. [Google Scholar]
  25. Loeser, R.F.; Pacione, C.A.; Chubinskaya, S. The combination of insulin-like growth factor 1 and osteogenic protein 1 promotes increased survival of and matrix synthesis by normal and osteoarthritic human articular chondrocytes. Arthritis Rheum 2003, 48, 2188–2196. [Google Scholar]
  26. Shi, S.; Mercer, S.; Eckert, G.J.; Trippel, S.B. Growth factor regulation of growth factors in articular chondrocytes. J. Biol. Chem 2009, 284, 6697–6704. [Google Scholar]
  27. Finnson, K.W.; Chi, Y.; Bou-Gharios, G.; Leask, A.; Philip, A. TGF-b signaling in cartilage homeostasis and osteoarthritis. Front. Biosci 2012, 4, 251–268. [Google Scholar]
  28. Guo, X.; Wang, X.F. Signaling cross-talk between TGF-β/BMP and other pathways. Cell Res 2009, 19, 71–88. [Google Scholar]
  29. Serra, R.; Johnson, M.; Filvaroff, E.H.; LaBorde, J.; Sheehan, D.M.; Derynck, R.; Moses, H.L. Expression of a truncated, kinase-defective TGF-β type II receptor in mouse skeletal tissue promotes terminal chondrocyte differentiation and osteoarthritis. J. Cell Biol 1997, 139, 541–552. [Google Scholar]
  30. Yang, X.; Chen, L.; Xu, X.; Li, C.; Huang, C.; Deng, C.X. TGF-beta/Smad3 signals repress chondrocyte hypertrophic differentiation and are required for maintaining articular cartilage. J. Cell Biol 2001, 153, 35–46. [Google Scholar]
  31. Van Beuningen, H.M.; van der Kraan, P.M.; Arntz, O.J.; van den Berg, W.B. Protection from interleukin 1 induced destruction of articular cartilage by transforming growth factor beta: Studies in anatomically intact cartilage in vitro and in vivo. Ann. Rheum. Dis 1993, 52, 185–191. [Google Scholar]
  32. Madry, H.; Kaul, G.; Cucchiarini, M.; Stein, U.; Zurakowski, D.; Remberger, K.; Menger, M.D.; Kohn, D.; Trippel, S.B. Enhanced repair of articular cartilage defects in vivo by transplanted chondrocytes overexpressing insulin-like growth factor I (IGF-I). Gene. Ther 2005, 12, 1171–1179. [Google Scholar]
  33. Chia, S.L.; Sawaji, Y.; Burleigh, A.; McLean, C.; Inglis, J.; Saklatvala, J.; Vincent, T. Fibroblast growth factor 2 is an intrinsic chondroprotective agent that suppresses ADAMTS-5 and delays cartilage degradation in murine osteoarthritis. Arthritis Rheum 2009, 60, 2019–2027. [Google Scholar]
  34. Hunter, D.J.; Pike, M.C.; Jonas, B.L.; Kissin, E.; Krop, J.; McAlindon, T. Phase 1 safety and tolerability study of BMP-7 in symptomatic knee osteoarthritis. BMC Musculoskelet. Disord 2010, 11. [Google Scholar] [CrossRef]
  35. Blaney Davidson, E.N.; Remst, D.F.; Vitters, E.L.; van Beuningen, H.M.; Blom, A.B.; Goumans, M.J.; van den Berg, W.B.; van der Kraan, P.M. Increase in ALK1/ALK5 ratio as a cause for elevated MMP-13 expression in osteoarthritis in humans and mice. J. Immunol 2009, 182, 7937–7945. [Google Scholar]
  36. Plaas, A.; Velasco, J.; Gorski, D.J.; Li, J.; Cole, A.; Christopherson, K.; Sandy, J.D. The relationship between fibrogenic TGFbeta1 signaling in the joint and cartilage degradation in post-injury osteoarthritis. Osteoarthr. Cartil 2011, 19, 1081–1090. [Google Scholar]
  37. Zhen, G.; Wen, C.; Jia, X.; Li, Y.; Crane, J.L.; Mears, S.C.; Askin, F.B.; Frassica, F.J.; Chang, W.; Yao, J.; et al. Inhibition of TGF-β signaling in mesenchymal stem cells of subchondral bone attenuates osteoarthritis. Nat. Med 2013, 19, 704–712. [Google Scholar]
  38. Konig, H.G.; Kogel, D.; Rami, A.; Prehn, J.H. TGF-{beta}1 activates two distinct type I receptors in neurons: Implications for neuronal NF-{kappa}B signaling. J. Cell Biol 2005, 168, 1077–1086. [Google Scholar]
  39. Finnson, K.W.; Parker, W.L.; ten Dijke, P.; Thorikay, M.; Philip, A. ALK1 opposes ALK5/Smad3 signaling and expression of extracellular matrix components in human chondrocytes. J. Bone Miner. Res 2008, 23, 896–906. [Google Scholar]
  40. Goumans, M.J.; Valdimarsdottir, G.; Itoh, S.; Lebrin, F.; Larsson, J.; Mummery, C.; Karlsson, S.; ten Dijke, P. Activin receptor-like kinase (ALK)1 is an antagonistic mediator of lateral TGFbeta/ALK5 signaling. Mol. Cell 2003, 12, 817–828. [Google Scholar]
  41. Blaney Davidson, E.N.; Vitters, E.L.; van der Kraan, P.M.; van den Berg, W.B. Expression of transforming growth factor-beta (TGFbeta) and the TGFbeta signalling molecule SMAD-2P in spontaneous and instability-induced osteoarthritis: Role in cartilage degradation, chondrogenesis and osteophyte formation. Ann. Rheum. Dis 2006, 65, 1414–1421. [Google Scholar]
  42. Hellingman, C.A.; Davidson, E.N.; Koevoet, W.; Vitters, E.L.; van den Berg, W.B.; van Osch, G.J.; van der Kraan, P.M. Smad signaling determines chondrogenic differentiation of bone-marrow-derived mesenchymal stem cells: Inhibition of Smad1/5/8P prevents terminal differentiation and calcification. Tissue Eng. Part A 2011, 17, 1157–1167. [Google Scholar]
  43. Li, T.F.; Darowish, M.; Zuscik, M.J.; Chen, D.; Schwarz, E.M.; Rosier, R.N.; Drissi, H.; O'Keefe, R.J. Smad3-deficient chondrocytes have enhanced BMP signaling and accelerated differentiation. J. Bone Miner. Res 2006, 21, 4–16. [Google Scholar]
  44. Bertrand, J.; Cromme, C.; Umlauf, D.; Frank, S.; Pap, T. Molecular mechanisms of cartilage remodelling in osteoarthritis. Int. J. Biochem. Cell Biol 2010, 42, 1594–1601. [Google Scholar]
  45. Van Donkelaar, C.C.; Wilson, W. Mechanics of chondrocyte hypertrophy. Biomech. Model. Mechanobiol 2011, 11, 655–664. [Google Scholar]
  46. Van der Kraan, P.M.; Blaney Davidson, E.N.; van den Berg, W.B. Bone morphogenetic proteins and articular cartilage: To serve and protect or a wolf in sheep clothing’s? Osteoarthr. Cartil 2010, 18, 735–741. [Google Scholar]
  47. Nishimura, R.; Hata, K.; Matsubara, T.; Wakabayashi, M.; Yoneda, T. Regulation of bone and cartilage development by network between BMP signalling and transcription factors. J. Biochem 2012, 151, 247–254. [Google Scholar]
  48. Chen, A.L.; Fang, C.; Liu, C.; Leslie, M.P.; Chang, E.; di Cesare, P.E. Expression of bone morphogenetic proteins, receptors, and tissue inhibitors in human fetal, adult, and osteoarthritic articular cartilage. J. Orthop. Res 2004, 22, 1188–1192. [Google Scholar]
  49. Minina, E.; Wenzel, H.M.; Kreschel, C.; Karp, S.; Gaffield, W.; McMahon, A.P.; Vortkamp, A. BMP and Ihh/PTHrP signaling interact to coordinate chondrocyte proliferation and differentiation. Development 2001, 128, 4523–4534. [Google Scholar]
  50. Chubinskaya, S.; Segalite, D.; Pikovsky, D.; Hakimiyan, A.A.; Rueger, D.C. Effects induced by BMPS in cultures of human articular chondrocytes: Comparative studies. Growth Factors 2008, 26, 275–283. [Google Scholar]
  51. Goldring, M.B.; Tsuchimochi, K.; Ijiri, K. The control of chondrogenesis. J. Cell Biochem 2006, 97, 33–44. [Google Scholar]
  52. Blaney Davidson, E.N.; Vitters, E.L.; van Lent, P.L.; van de Loo, F.A.; van den Berg, W.B.; van der Kraan, P.M. Elevated extracellular matrix production and degradation upon bone morphogenetic protein-2 (BMP-2) stimulation point toward a role for BMP-2 in cartilage repair and remodeling. Arthritis Res. Ther 2007, 9, R102. [Google Scholar]
  53. Zhao, L.; Li, G.; Zhou, G.Q. SOX9 directly binds CREB as a novel synergism with the PKA pathway in BMP-2-induced osteochondrogenic differentiation. J. Bone Miner. Res 2009, 24, 826–836. [Google Scholar]
  54. Pan, Q.; Yu, Y.; Chen, Q.; Li, C.; Wu, H.; Wan, Y.; Ma, J.; Sun, F. Sox9, a key transcription factor of bone morphogenetic protein-2-induced chondrogenesis, is activated through BMP pathway and a CCAAT box in the proximal promoter. J. Cell. Physiol 2008, 217, 228–241. [Google Scholar]
  55. Chimal-Monroy, J.; Rodriguez-Leon, J.; Montero, J.A.; Ganan, Y.; Macias, D.; Merino, R.; Hurle, J.M. Analysis of the molecular cascade responsible for mesodermal limb chondrogenesis: Sox genes and BMP signaling. Dev. Biol 2003, 257, 292–301. [Google Scholar]
  56. Lories, R.J.; Daans, M.; Derese, I.; Matthys, P.; Kasran, A.; Tylzanowski, P.; Ceuppens, J.L.; Luyten, F.P. Noggin haploinsufficiency differentially affects tissue responses in destructive and remodeling arthritis. Arthritis Rheum 2006, 54, 1736–1746. [Google Scholar]
  57. Papathanasiou, I.; Malizos, K.N.; Tsezou, A. Bone morphogenetic protein-2-induced Wnt/beta-catenin signaling pathway activation through enhanced low-density-lipoprotein receptor-related protein 5 catabolic activity contributes to hypertrophy in osteoarthritic chondrocytes. Arthritis Res. Ther 2012, 14, R82. [Google Scholar]
  58. Caron, M.M.; Emans, P.J.; Cremers, A.; Surtel, D.A.; Coolsen, M.M.; van Rhijn, L.W.; Welting, T.J. Hypertrophic differentiation during chondrogenic differentiation of progenitor cells is stimulated by BMP-2 but suppressed by BMP-7. Osteoarthr. Cartil 2013, 21, 604–613. [Google Scholar]
  59. Horiki, M.; Imamura, T.; Okamoto, M.; Hayashi, M.; Murai, J.; Myoui, A.; Ochi, T.; Miyazono, K.; Yoshikawa, H.; Tsumaki, N. Smad6/Smurf1 overexpression in cartilage delays chondrocyte hypertrophy and causes dwarfism with osteopenia. J. Cell Biol 2004, 165, 433–445. [Google Scholar]
  60. Retting, K.N.; Song, B.; Yoon, B.S.; Lyons, K.M. BMP canonical Smad signaling through Smad1 and Smad5 is required for endochondral bone formation. Development 2009, 136, 1093–1104. [Google Scholar]
  61. Fan, Z.; Chubinskaya, S.; Rueger, D.C.; Bau, B.; Haag, J.; Aigner, T. Regulation of anabolic and catabolic gene expression in normal and osteoarthritic adult human articular chondrocytes by osteogenic protein-1. Clin. Exp. Rheumatol 2004, 22, 103–106. [Google Scholar]
  62. Flechtenmacher, J.; Huch, K.; Thonar, E.J.; Mollenhauer, J.A.; Davies, S.R.; Schmid, T.M.; Puhl, W.; Sampath, T.K.; Aydelotte, M.B.; Kuettner, K.E. Recombinant human osteogenic protein 1 is a potent stimulator of the synthesis of cartilage proteoglycans and collagens by human articular chondrocytes. Arthritis Rheum 1996, 39, 1896–1904. [Google Scholar]
  63. Nishida, Y.; Knudson, C.B.; Knudson, W. Osteogenic Protein-1 inhibits matrix depletion in a hyaluronan hexasaccharide-induced model of osteoarthritis. Osteoarthr. Cartil 2004, 12, 374–382. [Google Scholar]
  64. Huch, K.; Wilbrink, B.; Flechtenmacher, J.; Koepp, H.E.; Aydelotte, M.B.; Sampath, T.K.; Kuettner, K.E.; Mollenhauer, J.; Thonar, E.J. Effects of recombinant human osteogenic protein 1 on the production of proteoglycan, prostaglandin E2, and interleukin-1 receptor antagonist by human articular chondrocytes cultured in the presence of interleukin-1beta. Arthritis Rheum 1997, 40, 2157–2161. [Google Scholar]
  65. Koepp, H.E.; Sampath, K.T.; Kuettner, K.E.; Homandberg, G.A. Osteogenic protein-1 (OP-1) blocks cartilage damage caused by fibronectin fragments and promotes repair by enhancing proteoglycan synthesis. Inflamm. Res 1999, 48, 199–204. [Google Scholar]
  66. Umulis, D.; O’Connor, M.B.; Blair, S.S. The extracellular regulation of bone morphogenetic protein signaling. Development 2009, 136, 3715–3728. [Google Scholar]
  67. Miyazono, K.; Kamiya, Y.; Morikawa, M. Bone morphogenetic protein receptors and signal transduction. J. Biochem 2010, 147, 35–51. [Google Scholar]
  68. Andhare, R.A.; Takahashi, N.; Knudson, W.; Knudson, C.B. Hyaluronan promotes the chondrocyte response to BMP-7. Osteoarthr. Cartil 2009, 17, 906–916. [Google Scholar]
  69. Luo, N.; Knudson, W.; Askew, E.B.; Veluci, R.; Knudson, C.B. CD44 and hyaluronan promote the BMP7 signaling response in chondrocytes. Arthritis Rheumatol 2014. [Google Scholar] [CrossRef]
  70. Kanakaris, N.K.; Calori, G.M.; Verdonk, R.; Burssens, P.; de Biase, P.; Capanna, R.; Vangosa, L.B.; Cherubino, P.; Baldo, F.; Ristiniemi, J.; et al. Application of BMP-7 to tibial non-unions: A 3-year multicenter experience. Injury 2008, 39, S83–S90. [Google Scholar]
  71. Carreira, A.C.; Lojudice, F.H.; Halcsik, E.; Navarro, R.D.; Sogayar, M.C.; Granjeiro, J.M. Bone Morphogenetic Proteins: Facts, Challenges, and Future Perspectives. J. Dent. Res 2014, 93, 335–345. [Google Scholar]
  72. Bessa, P.C.; Casal, M.; Reis, R.L. Bone morphogenetic proteins in tissue engineering: The road from laboratory to clinic, part II (BMP delivery). J. Tissue Eng. Regen. Med 2008, 2, 81–96. [Google Scholar]
  73. Posever, J.; Phillips, F.M.; Pottenger, L.A. Effects of basic fibroblast growth factor, transforming growth factor-beta 1, insulin-like growth factor-1, and insulin on human osteoarthritic articular cartilage explants. J. Orthop. Res 1995, 13, 832–837. [Google Scholar]
  74. Wang, E.; Wang, J.; Chin, E.; Zhou, J.; Bondy, C.A. Cellular patterns of insulin-like growth factor system gene expression in murine chondrogenesis and osteogenesis. Endocrinology 1995, 136, 2741–2751. [Google Scholar]
  75. Middleton, J.; Manthey, A.; Tyler, J. Insulin-like growth factor (IGF) receptor, IGF-I, interleukin-1 beta (IL-1 beta), and IL-6 mRNA expression in osteoarthritic and normal human cartilage. J. Histochem. Cytochem 1996, 44, 133–141. [Google Scholar]
  76. Sah, R.L.; Chen, A.C.; Grodzinsky, A.J.; Trippel, S.B. Differential effects of bFGF and IGF-I on matrix metabolism in calf and adult bovine cartilage explants. Arch. Biochem. Biophys 1994, 308, 137–147. [Google Scholar]
  77. Fukumoto, T.; Sperling, J.W.; Sanyal, A.; Fitzsimmons, J.S.; Reinholz, G.G.; Conover, C.A.; O’Driscoll, S.W. Combined effects of insulin-like growth factor-1 and transforming growth factor-beta1 on periosteal mesenchymal cells during chondrogenesis in vitro. Osteoarthr. Cartil. 2003, 11, 55–64. [Google Scholar]
  78. McQuillan, D.J.; Handley, C.J.; Campbell, M.A.; Bolis, S.; Milway, V.E.; Herington, A.C. Stimulation of proteoglycan biosynthesis by serum and insulin-like growth factor-I in cultured bovine articular cartilage. Biochem. J 1986, 240, 423–430. [Google Scholar]
  79. Luyten, F.P.; Hascall, V.C.; Nissley, S.P.; Morales, T.I.; Reddi, A.H. Insulin-like growth factors maintain steady-state metabolism of proteoglycans in bovine articular cartilage explants. Arch. Biochem. Biophys 1988, 267, 416–425. [Google Scholar]
  80. Maor, G.; Hochberg, Z.; Silbermann, M. Insulin-like growth factor I accelerates proliferation and differentiation of cartilage progenitor cells in cultures of neonatal mandibular condyles. Acta Endocrinol 1993, 128, 56–64. [Google Scholar]
  81. Bohme, K.; Conscience-Egli, M.; Tschan, T.; Winterhalter, K.H.; Bruckner, P. Induction of proliferation or hypertrophy of chondrocytes in serum-free culture: The role of insulin-like growth factor-I, insulin, or thyroxine. J. Cell Biol 1992, 116, 1035–1042. [Google Scholar]
  82. Ekenstedt, K.J.; Sonntag, W.E.; Loeser, R.F.; Lindgren, B.R.; Carlson, C.S. Effects of chronic growth hormone and insulin-like growth factor 1 deficiency on osteoarthritis severity in rat knee joints. Arthritis Rheum 2006, 54, 3850–3858. [Google Scholar]
  83. Fortier, L.A.; Mohammed, H.O.; Lust, G.; Nixon, A.J. Insulin-like growth factor-I enhances cell-based repair of articular cartilage. J. Bone Jt. Surg. Br 2002, 84, 276–288. [Google Scholar]
  84. Goodrich, L.R.; Hidaka, C.; Robbins, P.D.; Evans, C.H.; Nixon, A.J. Genetic modification of chondrocytes with insulin-like growth factor-1 enhances cartilage healing in an equine model. J. Bone Jt. Surg. Br 2007, 89, 672–685. [Google Scholar]
  85. Rosselot, G.; Vasilatos-Younken, R.; Leach, R.M. Effect of growth hormone, insulin-like growth factor I, basic fibroblast growth factor, and transforming growth factor beta on cell proliferation and proteoglycan synthesis by avian postembryonic growth plate chondrocytes. J. Bone Miner. Res 1994, 9, 431–439. [Google Scholar]
  86. Tsukazaki, T.; Usa, T.; Matsumoto, T.; Enomoto, H.; Ohtsuru, A.; Namba, H.; Iwasaki, K.; Yamashita, S. Effect of transforming growth factor-beta on the insulin-like growth factor-I autocrine/paracrine axis in cultured rat articular chondrocytes. Exp. Cell Res 1994, 215, 9–16. [Google Scholar]
  87. Yaeger, P.C.; Masi, T.L.; de Ortiz, J.L.; Binette, F.; Tubo, R.; McPherson, J.M. Synergistic action of transforming growth factor-beta and insulin-like growth factor-I induces expression of type II collagen and aggrecan genes in adult human articular chondrocytes. Exp. Cell Res 1997, 237, 318–325. [Google Scholar]
  88. Longobardi, L.; O’Rear, L.; Aakula, S.; Johnstone, B.; Shimer, K.; Chytil, A.; Horton, W.A.; Moses, H.L.; Spagnoli, A. Effect of IGF-I in the chondrogenesis of bone marrow mesenchymal stem cells in the presence or absence of TGF-beta signaling. J. Bone Miner. Res 2006, 21, 626–636. [Google Scholar]
  89. Worster, A.A.; Brower-Toland, B.D.; Fortier, L.A.; Bent, S.J.; Williams, J.; Nixon, A.J. Chondrocytic differentiation of mesenchymal stem cells sequentially exposed to transforming growth factor-beta1 in monolayer and insulin-like growth factor-I in a three-dimensional matrix. J. Orthop. Res 2001, 19, 738–749. [Google Scholar]
  90. Fortier, L.A.; Miller, B.J. Signaling through the small G-protein Cdc42 is involved in insulin-like growth factor-I resistance in aging articular chondrocytes. J. Orthop. Res 2006, 24, 1765–1772. [Google Scholar]
  91. Loeser, R.F.; Carlson, C.S.; del Carlo, M.; Cole, A. Detection of nitrotyrosine in aging and osteoarthritic cartilage: Correlation of oxidative damage with the presence of interleukin-1beta and with chondrocyte resistance to insulin-like growth factor 1. Arthritis Rheum 2002, 46, 2349–2357. [Google Scholar]
  92. Martin, J.A.; Ellerbroek, S.M.; Buckwalter, J.A. Age-related decline in chondrocyte response to insulin-like growth factor-I: The role of growth factor binding proteins. J. Orthop. Res 1997, 15, 491–498. [Google Scholar]
  93. Dore, S.; Pelletier, J.P.; DiBattista, J.A.; Tardif, G.; Brazeau, P.; Martel-Pelletier, J. Human osteoarthritic chondrocytes possess an increased number of insulin-like growth factor 1 binding sites but are unresponsive to its stimulation. Possible role of IGF-1-binding proteins. Arthritis Rheum 1994, 37, 253–263. [Google Scholar]
  94. Loeser, R.F.; Shanker, G.; Carlson, C.S.; Gardin, J.F.; Shelton, B.J.; Sonntag, W.E. Reduction in the chondrocyte response to insulin-like growth factor 1 in aging and osteoarthritis: Studies in a non-human primate model of naturally occurring disease. Arthritis Rheum 2000, 43, 2110–2120. [Google Scholar]
  95. Morales, T.I. The quantitative and functional relation between insulin-like growth factor-I (IGF) and IGF-binding proteins during human osteoarthritis. J. Orthop. Res 2008, 26, 465–474. [Google Scholar]
  96. Chubinskaya, S.; Hakimiyan, A.; Pacione, C.; Yanke, A.; Rappoport, L.; Aigner, T.; Rueger, D.C.; Loeser, R.F. Synergistic effect of IGF-1 and OP-1 on matrix formation by normal and OA chondrocytes cultured in alginate beads. Osteoarthr. Cartil 2007, 15, 421–430. [Google Scholar]
  97. Ellman, M.B.; An, H.S.; Muddasani, P.; Im, H.J. Biological impact of the fibroblast growth factor family on articular cartilage and intervertebral disc homeostasis. Gene 2008, 420, 82–89. [Google Scholar]
  98. Fortier, L.A.; Barker, J.U.; Strauss, E.J.; McCarrel, T.M.; Cole, B.J. The role of growth factors in cartilage repair. Clin. Orthop. Relat. Res 2011, 469, 2706–2715. [Google Scholar]
  99. Hiraide, A.; Yokoo, N.; Xin, K.Q.; Okuda, K.; Mizukami, H.; Ozawa, K.; Saito, T. Repair of articular cartilage defect by intraarticular administration of basic fibroblast growth factor gene, using adeno-associated virus vector. Hum. Gene Ther 2005, 16, 1413–1421. [Google Scholar]
  100. Deng, T.; Huang, S.; Zhou, S.; He, L.; Jin, Y. Cartilage regeneration using a novel gelatin-chondroitin-hyaluronan hybrid scaffold containing bFGF-impregnated microspheres. J. Microencapsul 2007, 24, 163–174. [Google Scholar]
  101. Inoue, A.; Takahashi, K.A.; Arai, Y.; Tonomura, H.; Sakao, K.; Saito, M.; Fujioka, M.; Fujiwara, H.; Tabata, Y.; Kubo, T. The therapeutic effects of basic fibroblast growth factor contained in gelatin hydrogel microspheres on experimental osteoarthritis in the rabbit knee. Arthritis Rheum 2006, 54, 264–270. [Google Scholar]
  102. Kaul, G.; Cucchiarini, M.; Arntzen, D.; Zurakowski, D.; Menger, M.D.; Kohn, D.; Trippel, S.B.; Madry, H. Local stimulation of articular cartilage repair by transplantation of encapsulated chondrocytes overexpressing human fibroblast growth factor 2 (FGF-2) in vivo. J. Gene Med. 2006, 8, 100–111. [Google Scholar]
  103. Stewart, K.; Pabbruwe, M.; Dickinson, S.; Sims, T.; Hollander, A.P.; Chaudhuri, J.B. The effect of growth factor treatment on meniscal chondrocyte proliferation and differentiation on polyglycolic acid scaffolds. Tissue Eng 2007, 13, 271–280. [Google Scholar]
  104. Loeser, R.F.; Chubinskaya, S.; Pacione, C.; Im, H.J. Basic fibroblast growth factor inhibits the anabolic activity of insulin-like growth factor 1 and osteogenic protein 1 in adult human articular chondrocytes. Arthritis Rheum 2005, 52, 3910–3917. [Google Scholar]
  105. Im, H.J.; Li, X.; Muddasani, P.; Kim, G.H.; Davis, F.; Rangan, J.; Forsyth, C.B.; Ellman, M.; Thonar, E.J. Basic fibroblast growth factor accelerates matrix degradation via a neuro-endocrine pathway in human adult articular chondrocytes. J. Cell. Physiol 2008, 215, 452–463. [Google Scholar]
  106. Im, H.J.; Muddasani, P.; Natarajan, V.; Schmid, T.M.; Block, J.A.; Davis, F.; van Wijnen, A.J.; Loeser, R.F. Basic fibroblast growth factor stimulates matrix metalloproteinase-13 via the molecular cross-talk between the mitogen-activated protein kinases and protein kinase Cdelta pathways in human adult articular chondrocytes. J. Biol. Chem 2007, 282, 11110–11121. [Google Scholar]
  107. Muddasani, P.; Norman, J.C.; Ellman, M.; van Wijnen, A.J.; Im, H.J. Basic fibroblast growth factor activates the MAPK and NFkappaB pathways that converge on Elk-1 to control production of matrix metalloproteinase-13 by human adult articular chondrocytes. J. Biol. Chem 2007, 282, 31409–31421. [Google Scholar]
  108. Ellsworth, J.L.; Berry, J.; Bukowski, T.; Claus, J.; Feldhaus, A.; Holderman, S.; Holdren, M.S.; Lum, K.D.; Moore, E.E.; Raymond, F.; et al. Fibroblast growth factor-18 is a trophic factor for mature chondrocytes and their progenitors. Osteoarthr. Cartil 2002, 10, 308–320. [Google Scholar]
  109. Davidson, D.; Blanc, A.; Filion, D.; Wang, H.; Plut, P.; Pfeffer, G.; Buschmann, M.D.; Henderson, J.E. Fibroblast growth factor (FGF) 18 signals through FGF receptor 3 to promote chondrogenesis. J. Biol. Chem 2005, 280, 20509–20515. [Google Scholar]
  110. Liu, Z.; Xu, J.; Colvin, J.S.; Ornitz, D.M. Coordination of chondrogenesis and osteogenesis by fibroblast growth factor 18. Genes Dev 2002, 16, 859–869. [Google Scholar]
  111. Moore, E.E.; Bendele, A.M.; Thompson, D.L.; Littau, A.; Waggie, K.S.; Reardon, B.; Ellsworth, J.L. Fibroblast growth factor-18 stimulates chondrogenesis and cartilage repair in a rat model of injury-induced osteoarthritis. Osteoarthr. Cartil 2005, 13, 623–631. [Google Scholar]
  112. Wang, G.L.; Jiang, B.H.; Rue, E.A.; Semenza, G.L. Hypoxia-inducible factor 1 is a basic-helix-loop-helix-PAS heterodimer regulated by cellular O2 tension. Proc. Natl. Acad. Sci. USA 1995, 92, 5510–5514. [Google Scholar]
  113. Semenza, G.L. HIF-1 and human disease: One highly involved factor. Genes Dev 2000, 14, 1983–1991. [Google Scholar]
  114. Schipani, E.; Ryan, H.E.; Didrickson, S.; Kobayashi, T.; Knight, M.; Johnson, R.S. Hypoxia in cartilage: HIF-1alpha is essential for chondrocyte growth arrest and survival. Genes Dev 2001, 15, 2865–2876. [Google Scholar]
  115. Forsythe, J.A.; Jiang, B.H.; Iyer, N.V.; Agani, F.; Leung, S.W.; Koos, R.D.; Semenza, G.L. Activation of vascular endothelial growth factor gene transcription by hypoxia-inducible factor 1. Mol. Cell Biol 1996, 16, 4604–4613. [Google Scholar]
  116. Murata, M.; Yudoh, K.; Masuko, K. The potential role of vascular endothelial growth factor (VEGF) in cartilage: How the angiogenic factor could be involved in the pathogenesis of osteoarthritis? Osteoarthr. Cartil 2008, 16, 279–286. [Google Scholar]
  117. Duval, E.; Leclercq, S.; Elissalde, J.M.; Demoor, M.; Galera, P.; Boumediene, K. Hypoxia-inducible factor 1alpha inhibits the fibroblast-like markers type I and type III collagen during hypoxia-induced chondrocyte redifferentiation: Hypoxia not only induces type II collagen and aggrecan, but it also inhibits type I and type III collagen in the hypoxia-inducible factor 1alpha-dependent redifferentiation of chondrocytes. Arthritis Rheum 2009, 60, 3038–3048. [Google Scholar]
  118. Aro, E.; Khatri, R.; Gerard-O’Riley, R.; Mangiavini, L.; Myllyharju, J.; Schipani, E. Hypoxia-inducible factor-1 (HIF-1) but not HIF-2 is essential for hypoxic induction of collagen prolyl 4-hydroxylases in primary newborn mouse epiphyseal growth plate chondrocytes. J. Biol. Chem 2012, 287, 37134–37144. [Google Scholar]
  119. Mobasheri, A.; Richardson, S.; Mobasheri, R.; Shakibaei, M.; Hoyland, J.A. Hypoxia inducible factor-1 and facilitative glucose transporters GLUT1 and GLUT3: Putative molecular components of the oxygen and glucose sensing apparatus in articular chondrocytes. Histol. Histopathol 2005, 20, 1327–1338. [Google Scholar]
  120. Pfander, D.; Cramer, T.; Swoboda, B. Hypoxia and HIF-1alpha in osteoarthritis. Int. Orthop 2005, 29, 6–9. [Google Scholar]
  121. Yudoh, K.; Nakamura, H.; Masuko-Hongo, K.; Kato, T.; Nishioka, K. Catabolic stress induces expression of hypoxia-inducible factor (HIF)-1 alpha in articular chondrocytes: Involvement of HIF-1 alpha in the pathogenesis of osteoarthritis. Arthritis Res. Ther 2005, 7, R904–R914. [Google Scholar]
  122. Aigner, T.; Zien, A.; Gehrsitz, A.; Gebhard, P.M.; McKenna, L. Anabolic and catabolic gene expression pattern analysis in normal versus osteoarthritic cartilage using complementary DNA-array technology. Arthritis Rheum 2001, 44, 2777–2789. [Google Scholar]
  123. Coimbra, I.B.; Jimenez, S.A.; Hawkins, D.F.; Piera-Velazquez, S.; Stokes, D.G. Hypoxia inducible factor-1 alpha expression in human normal and osteoarthritic chondrocytes. Osteoarthr. Cartil 2004, 12, 336–345. [Google Scholar]
  124. Stokes, D.G.; Liu, G.; Coimbra, I.B.; Piera-Velazquez, S.; Crowl, R.M.; Jimenez, S.A. Assessment of the gene expression profile of differentiated and dedifferentiated human fetal chondrocytes by microarray analysis. Arthritis Rheum 2002, 46, 404–419. [Google Scholar]
  125. Hellwig-Burgel, T.; Rutkowski, K.; Metzen, E.; Fandrey, J.; Jelkmann, W. Interleukin-1beta and tumor necrosis factor-alpha stimulate DNA binding of hypoxia-inducible factor-1. Blood 1999, 94, 1561–1567. [Google Scholar]
  126. Haddad, J.J.; Land, S.C. A non-hypoxic, ROS-sensitive pathway mediates TNF-alpha-dependent regulation of HIF-1alpha. FEBS Lett 2001, 505, 269–274. [Google Scholar]
  127. Haddad, J.J. Recombinant human interleukin (IL)-1 beta-mediated regulation of hypoxia-inducible factor-1 alpha (HIF-1 alpha) stabilization, nuclear translocation and activation requires an antioxidant/reactive oxygen species (ROS)-sensitive mechanism. Eur. Cytokine Netw 2002, 13, 250–260. [Google Scholar]
  128. Jung, Y.J.; Isaacs, J.S.; Lee, S.; Trepel, J.; Neckers, L. IL-1beta-mediated up-regulation of HIF-1alpha via an NFkappaB/COX-2 pathway identifies HIF-1 as a critical link between inflammation and oncogenesis. FASEB J 2003, 17, 2115–2117. [Google Scholar]
  129. Petersen, W.; Varoga, D.; Zantop, T.; Hassenpflug, J.; Mentlein, R.; Pufe, T. Cyclic strain influences the expression of the vascular endothelial growth factor (VEGF) and the hypoxia inducible factor 1 alpha (HIF-1alpha) in tendon fibroblasts. J. Orthop. Res 2004, 22, 847–853. [Google Scholar]
  130. Pufe, T.; Lemke, A.; Kurz, B.; Petersen, W.; Tillmann, B.; Grodzinsky, A.J.; Mentlein, R. Mechanical overload induces VEGF in cartilage discs via hypoxia-inducible factor. Am. J. Pathol 2004, 164, 185–192. [Google Scholar]
  131. Chang, H.; Shyu, K.G.; Wang, B.W.; Kuan, P. Regulation of hypoxia-inducible factor-1alpha by cyclical mechanical stretch in rat vascular smooth muscle cells. Clin. Sci 2003, 105, 447–456. [Google Scholar]
  132. Ema, M.; Taya, S.; Yokotani, N.; Sogawa, K.; Matsuda, Y.; Fujii-Kuriyama, Y. A novel bHLH-PAS factor with close sequence similarity to hypoxia-inducible factor 1alpha regulates the VEGF expression and is potentially involved in lung and vascular development. Proc. Natl. Acad. Sci. USA 1997, 94, 4273–4278. [Google Scholar]
  133. Patel, S.A.; Simon, M.C. Biology of hypoxia-inducible factor-2alpha in development and disease. Cell Death Differ 2008, 15, 628–634. [Google Scholar]
  134. Ratcliffe, P.J. HIF-1 and HIF-2: Working alone or together in hypoxia? J. Clin. Investig 2007, 117, 862–865. [Google Scholar]
  135. Wang, V.; Davis, D.A.; Haque, M.; Huang, L.E.; Yarchoan, R. Differential gene up-regulation by hypoxia-inducible factor-1alpha and hypoxia-inducible factor-2alpha in HEK293T cells. Cancer Res 2005, 65, 3299–3306. [Google Scholar]
  136. Lafont, J.E.; Talma, S.; Hopfgarten, C.; Murphy, C.L. Hypoxia promotes the differentiated human articular chondrocyte phenotype through SOX9-dependent and -independent pathways. J. Biol. Chem 2008, 283, 4778–4786. [Google Scholar]
  137. Lafont, J.E.; Talma, S.; Murphy, C.L. Hypoxia-inducible factor 2alpha is essential for hypoxic induction of the human articular chondrocyte phenotype. Arthritis Rheum 2007, 56, 3297–3306. [Google Scholar]
  138. Thoms, B.L.; Dudek, K.A.; Lafont, J.E.; Murphy, C.L. Hypoxia promotes the production and inhibits the destruction of human articular cartilage. Arthritis Rheum 2013, 65, 1302–1312. [Google Scholar]
  139. Saito, T.; Fukai, A.; Mabuchi, A.; Ikeda, T.; Yano, F.; Ohba, S.; Nishida, N.; Akune, T.; Yoshimura, N.; Nakagawa, T.; et al. Transcriptional regulation of endochondral ossification by HIF-2alpha during skeletal growth and osteoarthritis development. Nat. Med 2010, 16, 678–686. [Google Scholar]
  140. Kronenberg, H.M. Developmental regulation of the growth plate. Nature 2003, 423, 332–336. [Google Scholar]
  141. Mackie, E.J.; Ahmed, Y.A.; Tatarczuch, L.; Chen, K.S.; Mirams, M. Endochondral ossification: How cartilage is converted into bone in the developing skeleton. Int. J. Biochem. Cell Biol 2008, 40, 46–62. [Google Scholar]
  142. Tamiya, H.; Ikeda, T.; Jeong, J.H.; Saito, T.; Yano, F.; Jung, Y.K.; Ohba, S.; Kawaguchi, H.; Chung, U.I.; Choi, J.Y. Analysis of the Runx2 promoter in osseous and non-osseous cells and identification of HIF2A as a potent transcription activator. Gene 2008, 416, 53–60. [Google Scholar]
  143. Kawaguchi, H. Endochondral ossification signals in cartilage degradation during osteoarthritis progression in experimental mouse models. Mol. Cells 2008, 25, 1–6. [Google Scholar]
  144. Yang, S.; Kim, J.; Ryu, J.H.; Oh, H.; Chun, C.H.; Kim, B.J.; Min, B.H.; Chun, J.S. Hypoxia-inducible factor-2alpha is a catabolic regulator of osteoarthritic cartilage destruction. Nat. Med 2010, 16, 687–693. [Google Scholar]
  145. Mizushima, N.; Klionsky, D.J. Protein turnover via autophagy: Implications for metabolism. Annu. Rev. Nutr 2007, 27, 19–40. [Google Scholar]
  146. Uchiyama, Y.; Shibata, M.; Koike, M.; Yoshimura, K.; Sasaki, M. Autophagy-physiology and pathophysiology. Histochem. Cell Biol 2008, 129, 407–420. [Google Scholar]
  147. Mizushima, N. Physiological functions of autophagy. Curr. Top. Microbiol. Immunol 2009, 335, 71–84. [Google Scholar]
  148. Salminen, A.; Kauppinen, A.; Suuronen, T.; Kaarniranta, K.; Ojala, J. ER stress in Alzheimer’s disease: A novel neuronal trigger for inflammation and Alzheimer’s pathology. J. Neuroinflamm 2009, 6. [Google Scholar] [CrossRef]
  149. Austin, R.C. The unfolded protein response in health and disease. Antioxid. Redox Signal 2009, 11, 2279–2287. [Google Scholar]
  150. Bohensky, J.; Terkhorn, S.P.; Freeman, T.A.; Adams, C.S.; Garcia, J.A.; Shapiro, I.M.; Srinivas, V. Regulation of autophagy in human and murine cartilage: Hypoxia-inducible factor 2 suppresses chondrocyte autophagy. Arthritis Rheum 2009, 60, 1406–1415. [Google Scholar]
  151. Staines, K.A.; Macrae, V.E.; Farquharson, C. Cartilage development and degeneration: A Wnt Wnt situation. Cell Biochem. Funct 2012, 30, 633–642. [Google Scholar]
  152. Ma, B.; Landman, E.B.; Miclea, R.L.; Wit, J.M.; Robanus-Maandag, E.C.; Post, J.N.; Karperien, M. WNT signaling and cartilage: Of mice and men. Calcif. Tissue Int 2013, 92, 399–411. [Google Scholar]
  153. Sharma, A.R.; Jagga, S.; Lee, S.S.; Nam, J.S. Interplay between cartilage and subchondral bone contributing to pathogenesis of osteoarthritis. Int. J. Mol. Sci 2013, 14, 19805–19830. [Google Scholar]
  154. Sassi, N.; Laadhar, L.; Allouche, M.; Achek, A.; Kallel-Sellami, M.; Makni, S.; Sellami, S. WNT signaling and chondrocytes: From cell fate determination to osteoarthritis physiopathology. J. Recept. Signal Transduct. Res 2014, 34, 73–80. [Google Scholar]
  155. Bhanot, P.; Brink, M.; Samos, C.H.; Hsieh, J.C.; Wang, Y.; Macke, J.P.; Andrew, D.; Nathans, J.; Nusse, R. A new member of the frizzled family from Drosophila functions as a Wingless receptor. Nature 1996, 382, 225–230. [Google Scholar]
  156. Dann, C.E.; Hsieh, J.C.; Rattner, A.; Sharma, D.; Nathans, J.; Leahy, D.J. Insights into Wnt binding and signalling from the structures of two Frizzled cysteine-rich domains. Nature 2001, 412, 86–90. [Google Scholar]
  157. Aberle, H.; Bauer, A.; Stappert, J.; Kispert, A.; Kemler, R. beta-catenin is a target for the ubiquitin-proteasome pathway. EMBO J 1997, 16, 3797–3804. [Google Scholar]
  158. Monroe, D.G.; McGee-Lawrence, M.E.; Oursler, M.J.; Westendorf, J.J. Update on Wnt signaling in bone cell biology and bone disease. Gene 2012, 492, 1–18. [Google Scholar]
  159. Logan, C.Y.; Nusse, R. The Wnt signaling pathway in development and disease. Annu. Rev. Cell Dev. Biol 2004, 20, 781–810. [Google Scholar]
  160. Nusse, R. Wnt signaling in disease and in development. Cell Res 2005, 15, 28–32. [Google Scholar]
  161. Chun, J.S.; Oh, H.; Yang, S.; Park, M. Wnt signaling in cartilage development and degeneration. BMB Rep 2008, 41, 485–494. [Google Scholar]
  162. Nalesso, G.; Sherwood, J.; Bertrand, J.; Pap, T.; Ramachandran, M.; de Bari, C.; Pitzalis, C.; dell’accio, F. WNT-3A modulates articular chondrocyte phenotype by activating both canonical and noncanonical pathways. J. Cell Biol 2011, 193, 551–564. [Google Scholar] [Green Version]
  163. Yasuhara, R.; Ohta, Y.; Yuasa, T.; Kondo, N.; Hoang, T.; Addya, S.; Fortina, P.; Pacifici, M.; Iwamoto, M.; Enomoto-Iwamoto, M. Roles of beta-catenin signaling in phenotypic expression and proliferation of articular cartilage superficial zone cells. Lab. Investig 2011, 91, 1739–1752. [Google Scholar]
  164. Golovchenko, S.; Hattori, T.; Hartmann, C.; Gebhardt, M.; Gebhard, S.; Hess, A.; Pausch, F.; Schlund, B.; von der Mark, K. Deletion of beta catenin in hypertrophic growth plate chondrocytes impairs trabecular bone formation. Bone 2013, 55, 102–112. [Google Scholar]
  165. Dell’Accio, F.; de Bari, C.; El Tawil, N.M.; Barone, F.; Mitsiadis, T.A.; O’Dowd, J.; Pitzalis, C. Activation of WNT and BMP signaling in adult human articular cartilage following mechanical injury. Arthritis Res. Ther 2006, 8, R139. [Google Scholar]
  166. Dell’accio, F.; de Bari, C.; Eltawil, N.M.; Vanhummelen, P.; Pitzalis, C. Identification of the molecular response of articular cartilage to injury, by microarray screening: Wnt-16 expression and signaling after injury and in osteoarthritis. Arthritis Rheum 2008, 58, 1410–1421. [Google Scholar]
  167. Eltawil, N.M.; de Bari, C.; Achan, P.; Pitzalis, C.; dell’accio, F. A novel in vivo murine model of cartilage regeneration. Age and strain-dependent outcome after joint surface injury. Osteoarthr. Cartil 2009, 17, 695–704. [Google Scholar]
  168. Lodewyckx, L.; Cailotto, F.; Thysen, S.; Luyten, F.P.; Lories, R.J. Tight regulation of wingless-type signaling in the articular cartilage—Subchondral bone biomechanical unit: Transcriptomics in Frzb-knockout mice. Arthritis Res. Ther 2012, 14, R16. [Google Scholar]
  169. Miclea, R.L.; Siebelt, M.; Finos, L.; Goeman, J.J.; Lowik, C.W.; Oostdijk, W.; Weinans, H.; Wit, J.M.; Robanus-Maandag, E.C.; Karperien, M. Inhibition of Gsk3beta in cartilage induces osteoarthritic features through activation of the canonical Wnt signaling pathway. Osteoarthr. Cartil 2011, 19, 1363–1372. [Google Scholar]
  170. Zhu, M.; Tang, D.; Wu, Q.; Hao, S.; Chen, M.; Xie, C.; Rosier, R.N.; O’Keefe, R.J.; Zuscik, M.; Chen, D. Activation of beta-catenin signaling in articular chondrocytes leads to osteoarthritis-like phenotype in adult beta-catenin conditional activation mice. J. Bone Miner. Res 2009, 24, 12–21. [Google Scholar]
  171. Loughlin, J.; Dowling, B.; Chapman, K.; Marcelline, L.; Mustafa, Z.; Southam, L.; Ferreira, A.; Ciesielski, C.; Carson, D.A.; Corr, M. Functional variants within the secreted frizzled-related protein 3 gene are associated with hip osteoarthritis in females. Proc. Natl. Acad. Sci. USA 2004, 101, 9757–9762. [Google Scholar]
  172. Urano, T.; Shiraki, M.; Narusawa, K.; Usui, T.; Sasaki, N.; Hosoi, T.; Ouchi, Y.; Nakamura, T.; Inoue, S. Q89R polymorphism in the LDL receptor-related protein 5 gene is associated with spinal osteoarthritis in postmenopausal Japanese women. Spine 2007, 32, 25–29. [Google Scholar]
  173. Diarra, D.; Stolina, M.; Polzer, K.; Zwerina, J.; Ominsky, M.S.; Dwyer, D.; Korb, A.; Smolen, J.; Hoffmann, M.; Scheinecker, C.; et al. Dickkopf-1 is a master regulator of joint remodeling. Nat. Med 2007, 13, 156–163. [Google Scholar]
  174. Lane, N.E.; Nevitt, M.C.; Lui, L.Y.; de Leon, P.; Corr, M. Wnt signaling antagonists are potential prognostic biomarkers for the progression of radiographic hip osteoarthritis in elderly Caucasian women. Arthritis Rheum 2007, 56, 3319–3325. [Google Scholar]
  175. Honsawek, S.; Tanavalee, A.; Yuktanandana, P.; Ngarmukos, S.; Saetan, N.; Tantavisut, S. Dickkopf-1 (Dkk-1) in plasma and synovial fluid is inversely correlated with radiographic severity of knee osteoarthritis patients. BMC Musculoskelet. Disord 2010, 11, 257. [Google Scholar]
  176. Oeckinghaus, A.; Ghosh, S. The NF-kappaB family of transcription factors and its regulation. Cold Spring Harb. Perspect. Biol 2009, 1, a000034. [Google Scholar]
  177. Marcu, K.B.; Otero, M.; Olivotto, E.; Borzi, R.M.; Goldring, M.B. NF-kappaB signaling: Multiple angles to target OA. Curr. Drug Targets 2010, 11, 599–613. [Google Scholar]
  178. Rigoglou, S.; Papavassiliou, A.G. The NF-kappaB signalling pathway in osteoarthritis. Int. J. Biochem. Cell Biol 2013, 45, 2580–2584. [Google Scholar]
  179. Karin, M.; Greten, F.R. NF-kappaB: Linking inflammation and immunity to cancer development and progression. Nat. Rev. Immunol 2005, 5, 749–759. [Google Scholar]
  180. Hayden, M.S.; Ghosh, S. Shared principles in NF-kappaB signaling. Cell 2008, 132, 344–362. [Google Scholar]
  181. Ge, X.P.; Gan, Y.H.; Zhang, C.G.; Zhou, C.Y.; Ma, K.T.; Meng, J.H.; Ma, X.C. Requirement of the NF-kappaB pathway for induction of Wnt-5A by interleukin-1beta in condylar chondrocytes of the temporomandibular joint: Functional crosstalk between the Wnt-5A and NF-kappaB signaling pathways. Osteoarthr. Cartil 2011, 19, 111–117. [Google Scholar]
  182. Niederberger, E.; Geisslinger, G. The IKK-NF-kappaB pathway: A source for novel molecular drug targets in pain therapy? FASEB J 2008, 22, 3432–3442. [Google Scholar]
  183. Pomerantz, J.L.; Baltimore, D. Two pathways to NF-kappaB. Mol. Cell 2002, 10, 693–695. [Google Scholar]
  184. Bonizzi, G.; Karin, M. The two NF-kappaB activation pathways and their role in innate and adaptive immunity. Trends Immunol 2004, 25, 280–288. [Google Scholar]
  185. Karin, M.; Yamamoto, Y.; Wang, Q.M. The IKK NF-kappa B system: A treasure trove for drug development. Nat. Rev. Drug Discov 2004, 3, 17–26. [Google Scholar]
  186. Yamamoto, Y.; Gaynor, R.B. IkappaB kinases: Key regulators of the NF-kappaB pathway. Trends Biochem. Sci 2004, 29, 72–79. [Google Scholar]
  187. Yasuda, T. Activation of Akt leading to NF-kappaB up-regulation in chondrocytes stimulated with fibronectin fragment. Biomed. Res 2011, 32, 209–215. [Google Scholar]
  188. Ulivi, V.; Giannoni, P.; Gentili, C.; Cancedda, R.; Descalzi, F. p38/NF-κB-dependent expression of COX-2 during differentiation and inflammatory response of chondrocytes. J. Cell Biochem 2008, 104, 1393–1406. [Google Scholar]
  189. DeGroot, J.; Verzijl, N.; Wenting-van Wijk, M.J.; Jacobs, K.M.; van El, B.; van Roermund, P.M.; Bank, R.A.; Bijlsma, J.W.; TeKoppele, J.M.; Lafeber, F.P. Accumulation of advanced glycation end products as a molecular mechanism for aging as a risk factor in osteoarthritis. Arthritis Rheum 2004, 50, 1207–1215. [Google Scholar]
  190. Steenvoorden, M.M.; Huizinga, T.W.; Verzijl, N.; Bank, R.A.; Ronday, H.K.; Luning, H.A.; Lafeber, F.P.; Toes, R.E.; DeGroot, J. Activation of receptor for advanced glycation end products in osteoarthritis leads to increased stimulation of chondrocytes and synoviocytes. Arthritis Rheum 2006, 54, 253–263. [Google Scholar]
  191. Goldring, M.B.; Otero, M.; Plumb, D.A.; Dragomir, C.; Favero, M.; El Hachem, K.; Hashimoto, K.; Roach, H.I.; Olivotto, E.; Borzi, R.M.; et al. Roles of inflammatory and anabolic cytokines in cartilage metabolism: Signals and multiple effectors converge upon MMP-13 regulation in osteoarthritis. Eur. Cell Mater 2011, 21, 202–220. [Google Scholar]
  192. Hirata, M.; Kugimiya, F.; Fukai, A.; Saito, T.; Yano, F.; Ikeda, T.; Mabuchi, A.; Sapkota, B.R.; Akune, T.; Nishida, N.; et al. C/EBPbeta and RUNX2 cooperate to degrade cartilage with MMP-13 as the target and HIF-2alpha as the inducer in chondrocytes. Hum. Mol. Genet 2012, 21, 1111–1123. [Google Scholar]
  193. Grall, F.; Gu, X.; Tan, L.; Cho, J.Y.; Inan, M.S.; Pettit, A.R.; Thamrongsak, U.; Choy, B.K.; Manning, C.; Akbarali, Y.; et al. Responses to the proinflammatory cytokines interleukin-1 and tumor necrosis factor alpha in cells derived from rheumatoid synovium and other joint tissues involve nuclear factor kappaB-mediated induction of the Ets transcription factor ESE-1. Arthritis Rheum 2003, 48, 1249–1260. [Google Scholar]
  194. Peng, H.; Tan, L.; Osaki, M.; Zhan, Y.; Ijiri, K.; Tsuchimochi, K.; Otero, M.; Wang, H.; Choy, B.K.; Grall, F.T.; et al. ESE-1 is a potent repressor of type II collagen gene (COL2A1) transcription in human chondrocytes. J. Cell. Physiol 2008, 215, 562–573. [Google Scholar]
  195. Otero, M.; Plumb, D.A.; Tsuchimochi, K.; Dragomir, C.L.; Hashimoto, K.; Peng, H.; Olivotto, E.; Bevilacqua, M.; Tan, L.; Yang, Z.; et al. E74-like factor 3 (ELF3) impacts on matrix metalloproteinase 13 (MMP13) transcriptional control in articular chondrocytes under proinflammatory stress. J. Biol. Chem 2012, 287, 3559–3572. [Google Scholar]
  196. Merz, D.; Liu, R.; Johnson, K.; Terkeltaub, R. IL-8/CXCL8 and growth-related oncogene alpha/CXCL1 induce chondrocyte hypertrophic differentiation. J. Immunol 2003, 171, 4406–4415. [Google Scholar]
  197. Ijiri, K.; Zerbini, L.F.; Peng, H.; Otu, H.H.; Tsuchimochi, K.; Otero, M.; Dragomir, C.; Walsh, N.; Bierbaum, B.E.; Mattingly, D.; et al. Differential expression of GADD45beta in normal and osteoarthritic cartilage: Potential role in homeostasis of articular chondrocytes. Arthritis Rheum 2008, 58, 2075–2087. [Google Scholar]
  198. Husa, M.; Liu-Bryan, R.; Terkeltaub, R. Shifting HIFs in osteoarthritis. Nat. Med 2010, 16, 641–644. [Google Scholar]
  199. Scanzello, C.R.; Goldring, S.R. The role of synovitis in osteoarthritis pathogenesis. Bone 2012, 51, 249–257. [Google Scholar]
  200. Liu, S.C.; Hsu, C.J.; Chen, H.T.; Tsou, H.K.; Chuang, S.M.; Tang, C.H. CTGF increases IL-6 expression in human synovial fibroblasts through integrin-dependent signaling pathway. PLoS One 2012, 7, e51097. [Google Scholar]
  201. Hou, C.H.; Tang, C.H.; Hsu, C.J.; Hou, S.M.; Liu, J.F. CCN4 induces IL-6 production through alphavbeta5 receptor, PI3K, Akt, and NF-kappaB singling pathway in human synovial fibroblasts. Arthritis Res. Ther 2013, 15, R19. [Google Scholar]
  202. Krasnokutsky, S.; Attur, M.; Palmer, G.; Samuels, J.; Abramson, S.B. Current concepts in the pathogenesis of osteoarthritis. Osteoarthr. Cartil 2008, 16, S1–S3. [Google Scholar]
  203. Xing, L.; Carlson, L.; Story, B.; Tai, Z.; Keng, P.; Siebenlist, U.; Boyce, B.F. Expression of either NF-kappaB p50 or p52 in osteoclast precursors is required for IL-1-induced bone resorption. J. Bone Miner. Res 2003, 18, 260–269. [Google Scholar]
  204. Nakashima, T.; Takayanagi, H. The dynamic interplay between osteoclasts and the immune system. Arch. Biochem. Biophys 2008, 473, 166–171. [Google Scholar]
  205. Sagar, D.R.; Ashraf, S.; Xu, L.; Burston, J.J.; Menhinick, M.R.; Poulter, C.L.; Bennett, A.J.; Walsh, D.A.; Chapman, V. Osteoprotegerin reduces the development of pain behaviour and joint pathology in a model of osteoarthritis. Ann. Rheum. Dis 2013. [Google Scholar] [CrossRef]
  206. Castaneda, S.; Roman-Blas, J.A.; Largo, R.; Herrero-Beaumont, G. Subchondral bone as a key target for osteoarthritis treatment. Biochem. Pharmacol 2012, 83, 315–323. [Google Scholar]
  207. Kwan Tat, S.; Amiable, N.; Pelletier, J.P.; Boileau, C.; Lajeunesse, D.; Duval, N.; Martel-Pelletier, J. Modulation of OPG, RANK and RANKL by human chondrocytes and their implication during osteoarthritis. Rheumatology 2009, 48, 1482–1490. [Google Scholar]
  208. Trouvin, A.P.; Goeb, V. Receptor activator of nuclear factor-kappaB ligand and osteoprotegerin: Maintaining the balance to prevent bone loss. Clin. Interv. Aging 2010, 5, 345–354. [Google Scholar]
  209. Martin, T.J. Historically significant events in the discovery of RANK/RANKL/OPG. World J. Orthop 2013, 4, 186–197. [Google Scholar]
  210. Bobick, B.E.; Kulyk, W.M. Regulation of cartilage formation and maturation by mitogen-activated protein kinase signaling. Birth Defects Res. C Embryo Today 2008, 84, 131–154. [Google Scholar]
  211. Chang, L.; Karin, M. Mammalian MAP kinase signalling cascades. Nature 2001, 410, 37–40. [Google Scholar]
  212. Takebe, K.; Nishiyama, T.; Hayashi, S.; Hashimoto, S.; Fujishiro, T.; Kanzaki, N.; Kawakita, K.; Iwasa, K.; Kuroda, R.; Kurosaka, M. Regulation of p38 MAPK phosphorylation inhibits chondrocyte apoptosis in response to heat stress or mechanical stress. Int. J. Mol. Med 2011, 27, 329–335. [Google Scholar]
  213. Prasadam, I.; van Gennip, S.; Friis, T.; Shi, W.; Crawford, R.; Xiao, Y. ERK-1/2 and p38 in the regulation of hypertrophic changes of normal articular cartilage chondrocytes induced by osteoarthritic subchondral osteoblasts. Arthritis Rheum 2010, 62, 1349–1360. [Google Scholar] [Green Version]
  214. Thalhamer, T.; McGrath, M.A.; Harnett, M.M. MAPKs and their relevance to arthritis and inflammation. Rheumatology 2008, 47, 409–414. [Google Scholar]
  215. Kyriakis, J.M.; Banerjee, P.; Nikolakaki, E.; Dai, T.; Rubie, E.A.; Ahmad, M.F.; Avruch, J.; Woodgett, J.R. The stress-activated protein kinase subfamily of c-Jun kinases. Nature 1994, 369, 156–160. [Google Scholar]
  216. Raingeaud, J.; Gupta, S.; Rogers, J.S.; Dickens, M.; Han, J.; Ulevitch, R.J.; Davis, R.J. Pro-inflammatory cytokines and environmental stress cause p38 mitogen-activated protein kinase activation by dual phosphorylation on tyrosine and threonine. J. Biol. Chem 1995, 270, 7420–7426. [Google Scholar]
  217. Freshney, N.W.; Rawlinson, L.; Guesdon, F.; Jones, E.; Cowley, S.; Hsuan, J.; Saklatvala, J. Interleukin-1 activates a novel protein kinase cascade that results in the phosphorylation of Hsp27. Cell 1994, 78, 1039–1049. [Google Scholar]
  218. Berenbaum, F. Signaling transduction: Target in osteoarthritis. Curr. Opin. Rheumatol 2004, 16, 616–622. [Google Scholar]
  219. Tetsunaga, T.; Nishida, K.; Furumatsu, T.; Naruse, K.; Hirohata, S.; Yoshida, A.; Saito, T.; Ozaki, T. Regulation of mechanical stress-induced MMP-13 and ADAMTS-5 expression by RUNX-2 transcriptional factor in SW1353 chondrocyte-like cells. Osteoarthr. Cartil 2011, 19, 222–232. [Google Scholar]
  220. Thirunavukkarasu, K.; Pei, Y.; Moore, T.L.; Wang, H.; Yu, X.P.; Geiser, A.G.; Chandrasekhar, S. Regulation of the human ADAMTS-4 promoter by transcription factors and cytokines. Biochem. Biophys. Res. Commun 2006, 345, 197–204. [Google Scholar]
  221. Houard, X.; Goldring, M.B.; Berenbaum, F. Homeostatic mechanisms in articular cartilage and role of inflammation in osteoarthritis. Curr. Rheumatol. Rep 2013, 15. [Google Scholar] [CrossRef]
  222. Stanton, L.A.; Underhill, T.M.; Beier, F. MAP kinases in chondrocyte differentiation. Dev. Biol 2003, 263, 165–175. [Google Scholar]
  223. Greenblatt, M.B.; Shim, J.H.; Glimcher, L.H. Mitogen-activated protein kinase pathways in osteoblasts. Annu. Rev. Cell Dev. Biol 2013, 29, 63–79. [Google Scholar]
  224. Prasadam, I.; Friis, T.; Shi, W.; van Gennip, S.; Crawford, R.; Xiao, Y. Osteoarthritic cartilage chondrocytes alter subchondral bone osteoblast differentiation via MAPK signalling pathway involving ERK1/2. Bone 2010, 46, 226–235. [Google Scholar]
  225. Oh, C.D.; Chang, S.H.; Yoon, Y.M.; Lee, S.J.; Lee, Y.S.; Kang, S.S.; Chun, J.S. Opposing role of mitogen-activated protein kinase subtypes, erk-1/2 and p38, in the regulation of chondrogenesis of mesenchymes. J. Biol. Chem 2000, 275, 5613–5619. [Google Scholar]
  226. Sondergaard, B.C.; Schultz, N.; Madsen, S.H.; Bay-Jensen, A.C.; Kassem, M.; Karsdal, M.A. MAPKs are essential upstream signaling pathways in proteolytic cartilage degradation—Divergence in pathways leading to aggrecanase and MMP-mediated articular cartilage degradation. Osteoarthr. Cartil 2010, 18, 279–288. [Google Scholar]
  227. Goldring, M.B.; Otero, M.; Tsuchimochi, K.; Ijiri, K.; Li, Y. Defining the roles of inflammatory and anabolic cytokines in cartilage metabolism. Ann. Rheum. Dis 2008, 67, iii75–iii82. [Google Scholar]
  228. Wang, X.; Li, F.; Fan, C.; Wang, C.; Ruan, H. Effects and relationship of ERK1 and ERK2 in interleukin-1β-induced alterations in MMP3, MMP13, type II collagen and aggrecan expression in human chondrocytes. Int. J. Mol. Med 2011, 27, 583–589. [Google Scholar]
  229. Djouad, F.; Rackwitz, L.; Song, Y.; Janjanin, S.; Tuan, R.S. ERK1/2 activation induced by inflammatory cytokines compromises effective host tissue integration of engineered cartilage. Tissue Eng. Part A 2009, 15, 2825–2835. [Google Scholar]
  230. Chung, U.I.; Schipani, E.; McMahon, A.P.; Kronenberg, H.M. Indian hedgehog couples chondrogenesis to osteogenesis in endochondral bone development. J. Clin. Investig 2001, 107, 295–304. [Google Scholar]
  231. Mak, K.K.; Kronenberg, H.M.; Chuang, P.T.; Mackem, S.; Yang, Y. Indian hedgehog signals independently of PTHrP to promote chondrocyte hypertrophy. Development 2008, 135, 1947–1956. [Google Scholar]
  232. Maeda, Y.; Nakamura, E.; Nguyen, M.T.; Suva, L.J.; Swain, F.L.; Razzaque, M.S.; Mackem, S.; Lanske, B. Indian Hedgehog produced by postnatal chondrocytes is essential for maintaining a growth plate and trabecular bone. Proc. Natl. Acad. Sci. USA 2007, 104, 6382–6387. [Google Scholar]
  233. Kobayashi, T.; Soegiarto, D.W.; Yang, Y.; Lanske, B.; Schipani, E.; McMahon, A.P.; Kronenberg, H.M. Indian hedgehog stimulates periarticular chondrocyte differentiation to regulate growth plate length independently of PTHrP. J. Clin. Investig 2005, 115, 1734–1742. [Google Scholar]
  234. Vortkamp, A.; Lee, K.; Lanske, B.; Segre, G.V.; Kronenberg, H.M.; Tabin, C.J. Regulation of rate of cartilage differentiation by Indian hedgehog and PTH-related protein. Science 1996, 273, 613–622. [Google Scholar]
  235. Lanske, B.; Karaplis, A.C.; Lee, K.; Luz, A.; Vortkamp, A.; Pirro, A.; Karperien, M.; Defize, L.H.; Ho, C.; Mulligan, R.C.; et al. PTH/PTHrP receptor in early development and Indian hedgehog-regulated bone growth. Science 1996, 273, 663–666. [Google Scholar]
  236. Ng, T.C.; Chiu, K.W.; Rabie, A.B.; Hagg, U. Repeated mechanical loading enhances the expression of Indian hedgehog in condylar cartilage. Front. Biosci 2006, 11, 943–948. [Google Scholar]
  237. Wilson, C.W.; Chuang, P.T. Mechanism and evolution of cytosolic Hedgehog signal transduction. Development 2010, 137, 2079–2094. [Google Scholar]
  238. Corbit, K.C.; Aanstad, P.; Singla, V.; Norman, A.R.; Stainier, D.Y.; Reiter, J.F. Vertebrate Smoothened functions at the primary cilium. Nature 2005, 437, 1018–1021. [Google Scholar]
  239. Wuelling, M.; Vortkamp, A. Transcriptional networks controlling chondrocyte proliferation and differentiation during endochondral ossification. Pediatr. Nephrol 2010, 25, 625–631. [Google Scholar]
  240. Lin, A.C.; Seeto, B.L.; Bartoszko, J.M.; Khoury, M.A.; Whetstone, H.; Ho, L.; Hsu, C.; Ali, S.A.; Alman, B.A. Modulating hedgehog signaling can attenuate the severity of osteoarthritis. Nat. Med 2009, 15, 1421–1425. [Google Scholar]
  241. St-Jacques, B.; Hammerschmidt, M.; McMahon, A.P. Indian hedgehog signaling regulates proliferation and differentiation of chondrocytes and is essential for bone formation. Genes Dev 1999, 13, 2072–2086. [Google Scholar]
  242. Chen, X.; Macica, C.M.; Nasiri, A.; Broadus, A.E. Regulation of articular chondrocyte proliferation and differentiation by indian hedgehog and parathyroid hormone-related protein in mice. Arthritis Rheum 2008, 58, 3788–3797. [Google Scholar]
  243. Wei, F.; Zhou, J.; Wei, X.; Zhang, J.; Fleming, B.C.; Terek, R.; Pei, M.; Chen, Q.; Liu, T.; Wei, L. Activation of Indian hedgehog promotes chondrocyte hypertrophy and upregulation of MMP-13 in human osteoarthritic cartilage. Osteoarthr. Cartil 2012, 20, 755–763. [Google Scholar]
  244. Steinert, A.F.; Weissenberger, M.; Kunz, M.; Gilbert, F.; Ghivizzani, S.C.; Gobel, S.; Jakob, F.; Noth, U.; Rudert, M. Indian hedgehog gene transfer is a chondrogenic inducer of human mesenchymal stem cells. Arthritis Res. Ther 2012, 14, R168. [Google Scholar]
  245. Lin, L.; Shen, Q.; Xue, T.; Duan, X.; Fu, X.; Yu, C. Sonic hedgehog improves redifferentiation of dedifferentiated chondrocytes for articular cartilage repair. PLoS One 2014, 9, e88550. [Google Scholar]
  246. Kafienah, W.; Mistry, S.; Dickinson, S.C.; Sims, T.J.; Learmonth, I.; Hollander, A.P. Three-dimensional cartilage tissue engineering using adult stem cells from osteoarthritis patients. Arthritis Rheum 2007, 56, 177–187. [Google Scholar]
  247. Wu, L.; Huang, X.; Li, L.; Huang, H.; Xu, R.; Luyten, W. Insights on biology and pathology of HIF-1α/-2α, TGFβ/BMP, Wnt/β-catenin, and NF-κB pathways in osteoarthritis. Curr. Pharm. Des 2012, 18, 3293–3312. [Google Scholar]
  248. Van den Bosch, M.H.; Blom, A.B.; van Lent, P.L.; van Beuningen, H.M.; Blaney Davidson, E.N.; van der Kraan, P.M.; van den Berg, W.B. Canonical Wnt signaling skews TGF-β signaling in chondrocytes towards signaling via ALK1 and Smad 1/5/8. Cell Signal 2014, 26, 951–958. [Google Scholar]
  249. Mak, K.K.; Chen, M.H.; Day, T.F.; Chuang, P.T.; Yang, Y. Wnt/beta-catenin signaling interacts differentially with Ihh signaling in controlling endochondral bone and synovial joint formation. Development 2006, 133, 3695–3707. [Google Scholar]
  250. Dao, D.Y.; Jonason, J.H.; Zhang, Y.; Hsu, W.; Chen, D.; Hilton, M.J.; O’Keefe, R.J. Cartilage-specific β-catenin signaling regulates chondrocyte maturation, generation of ossification centers, and perichondrial bone formation during skeletal development. J. Bone Miner. Res 2012, 27, 1680–1694. [Google Scholar]
Figure 1. Schematic overview of signaling cross-talk among transforming growth factor-β (TGF-β), bone morphogenetic proteins (BMPs), hypoxia-related factors (HIF), Wnt/β-catenin, nuclear factor kappa B (NF-κB), mitogen-activated protein kinase (MAPK) and Indian hedgehog (Ihh) pathways.
Figure 1. Schematic overview of signaling cross-talk among transforming growth factor-β (TGF-β), bone morphogenetic proteins (BMPs), hypoxia-related factors (HIF), Wnt/β-catenin, nuclear factor kappa B (NF-κB), mitogen-activated protein kinase (MAPK) and Indian hedgehog (Ihh) pathways.
Ijms 15 08667f1
Table 1. Role of Wnt signaling during early stages of cartilage development.
Table 1. Role of Wnt signaling during early stages of cartilage development.
Promoting Effects
Wnt5aExpressed in perichondrium surrounding condensations
Recruits mesenchymal stem cells
Delays chondrocyte differentiation
Wnt5bExpressed in pre-hypertrophic chondrocytes and in the perichondrium
Promotes initial steps of chondrogenesis in micromass cultures by stimulating cartilage nodule formation
Delays terminal differentiation in vivo
β-catenin low levelExpressed in chondrogenic mesenchymal condensations
Induces expression of Sox9 and promotes chondrogenic differentiation
Dkk1sFRP1Promotes early chondrogenesis in human mesenchymal stem cells
Wif-1Expressed in mesenchyme surrounding cartilage elements and articular cartilage
Promotes chondrogenic differentiation by neutralizing Wnt3a mediated inhibition of chondrogenesis
Inhibitory Effects
Wnt1Inhibits cell condensation and thus cartilage formation
Wnt3aExpressed in early stages of chondrogenesis, decreased when chondrogenic differentiation proceeds
Increases self-renewal and decreases apoptosis of MSCs Stabilizes cell-cell adhesion through promoting sustained expression of N-cadherin and β-catenin
Blocks collagen II expression and proteoglycan deposition
Wnt4Expressed in developing joint interzone
Inhibits cell condensation and thus chondrogenesis condensation
Wnt6Inhibits chondrogenesis at an early stage prior to chondrogenic differentiation (up-stream of SOX-9)
Wnt7aExpressed in dorsal ectoderm in developing limb
Inhibits chondrogenic differentiation in vitro and in vivo mediated by MAPK and AP-1 signaling
Prolongs stabilization of N-cadherin
Blocks transition from the condensation state to the cartilaginous nodule formation
Wnt9a (14)Expressed in developing joint interzone
Expressed in synovium and joint capsule in the mature joint
Arrests or reverses chondrogenic differentiation
Blocks transition from the condensation state to the cartilaginous nodule formation
Fzl7Inhibits mesenchymal condensation at the pre-cartilage aggregate formation by suppressing N-cadherin expression
Table 2. Role of Wnt signaling during late stages of cartilage development and adulthood.
Table 2. Role of Wnt signaling during late stages of cartilage development and adulthood.
Promoting Effects
Wnt-4Expressed in periphery of joint interzone and hypertrophic chondrocytes
Accelerates chondrocyte terminal differentiation
Induces terminal differentiation of growth plate cartilage
Wnt8Promotes chondrocyte hypertrophy
β-catenin high levelIncreases cell hypertrophy through RUNX-2 or IHH signaling activation
Induces collagen X expression
Inhibitory Effects
Wnt-5aWnt-5bInhibits hypertrophic maturation of chondrocytes through NF-κB stimulation and RUNX-2 inhibition
FRZBExpressed in prechondrogenic mesenchymal condensations and in epiphyseal pre-articular chondrocytes
Blocks chondrocyte maturation and prevents endochondral ossification in vivo
Promotes glycosaminoglycan synthesis and expression of Sox9 and collagen II in vitro
DKK-1Expressed at sites of programmed cell death in apical ectodermal ridge
Expressed at higher level in articular cartilage than growth plate cartilage
Promotes glycosaminoglycan synthesis and expression of Sox9 and collagen II in vitro
Inhibits chondrocyte hypertrophy
Fzl1Fzl7Blocks/delays chondrocyte hypertrophy
sFRP-1sFRP-3Delays terminal differentiation of hypertrophic chondrocytes

Share and Cite

MDPI and ACS Style

Mariani, E.; Pulsatelli, L.; Facchini, A. Signaling Pathways in Cartilage Repair. Int. J. Mol. Sci. 2014, 15, 8667-8698. https://doi.org/10.3390/ijms15058667

AMA Style

Mariani E, Pulsatelli L, Facchini A. Signaling Pathways in Cartilage Repair. International Journal of Molecular Sciences. 2014; 15(5):8667-8698. https://doi.org/10.3390/ijms15058667

Chicago/Turabian Style

Mariani, Erminia, Lia Pulsatelli, and Andrea Facchini. 2014. "Signaling Pathways in Cartilage Repair" International Journal of Molecular Sciences 15, no. 5: 8667-8698. https://doi.org/10.3390/ijms15058667

Article Metrics

Back to TopTop