Download as pdf or txt
Download as pdf or txt
You are on page 1of 18

Connective Tissue Research

ISSN: 0300-8207 (Print) 1607-8438 (Online) Journal homepage: http://www.tandfonline.com/loi/icts20

Recent advances in the understanding of


molecular mechanisms of cartilage degeneration,
synovitis and subchondral bone changes in
osteoarthritis

Yingliang Wei & Lunhao Bai

To cite this article: Yingliang Wei & Lunhao Bai (2016) Recent advances in the understanding
of molecular mechanisms of cartilage degeneration, synovitis and subchondral
bone changes in osteoarthritis, Connective Tissue Research, 57:4, 245-261, DOI:
10.1080/03008207.2016.1177036

To link to this article: http://dx.doi.org/10.1080/03008207.2016.1177036

Published online: 10 Jun 2016.

Submit your article to this journal

Article views: 400

View related articles

View Crossmark data

Full Terms & Conditions of access and use can be found at


http://www.tandfonline.com/action/journalInformation?journalCode=icts20

Download by: [200.123.111.226] Date: 27 September 2016, At: 04:42


CONNECTIVE TISSUE RESEARCH
2016, VOL. 57, NO. 4, 245–261
http://dx.doi.org/10.1080/03008207.2016.1177036

REVIEW

Recent advances in the understanding of molecular mechanisms of cartilage


degeneration, synovitis and subchondral bone changes in osteoarthritis
Yingliang Wei and Lunhao Bai
Department of Orthopedic Surgery, Sheng–Jing Hospital, China Medical University, ShenYang, China

ABSTRACT ARTICLE HISTORY


Osteoarthritis (OA), the most common form of degenerative joint disease, is linked to high Received 26 October 2015
morbidity. It is predicted to be the single greatest cause of disability in the general population Revised 22 March 2016
by 2030. The development of disease-modifying therapy for OA currently face great obstacle Accepted 5 April 2016
Published online 9 June 2016
mainly because the onset and development of the disease involve complex molecular mechan-
isms. In this review, we will comprehensively summarize biological and pathological mechanisms KEYWORDS
of three key aspects: degeneration of articular cartilage, synovial immunopathogenesis, and Cartilage degeneration;
changes in subchondral bone. For each tissue, we will focus on the molecular receptors, cytokines, inflammation; molecular
peptidases, related cell, and signal pathways. Agents that specifically block mechanisms involved mechanisms; osteoarthritis;
in synovial inflammation, degeneration of articular cartilage, and subchondral bone remodeling subchondral bone
can potentially be exploited to produce targeted therapy for OA. Such new comprehensive agents
will benefit affected patients and bring exciting new hope for the treatment of OA.

Introduction calcified cartilage zone (6). The chondrocytes are


embedded in a matrix consisting of a network of col-
Osteoarthritis (OA), the most common form of degen-
lagen fibers (primarily type II) as well as varying
erative joint disease, is a major health burden as it is the
amounts of proteoglycans. The chondrocytes produce
most common form of arthritis and is linked to high
and maintain the physical function of cartilage by
morbidity. It is also predicted to be the single greatest
synthesizing and degrading matrix components in
cause of disability in the general population by 2030 (1),
response to environmental changes such as changes in
with an estimated 35% of people eventually suffering from
growth factors, cytokines, and biomechanical forces
OA. Disease rate is likely to increase year by year con-
(7,8). Once the microenvironment changes, the altera-
tinuously as the global population ages and working lives
tion in the normal physiologic balance under the influ-
become longer in developed countries (2). Despite its high
ence of all kinds of factors lead to functional
prevalence and substantial public health impact, disease
abnormality of these cells. However, the precise mole-
etiology remains incompletely understood. While several
cular mechanism initiating chondrocyte malfunction-
risk factors have been associated with OA, including
ing remains unknown. In this review, we discuss recent
genetic predisposition, aging, obesity, and joint malalign-
molecular mechanisms involved in the development of
ment (3,4), OA is not a simple process of wear and tear
OA. Our review will focus on receptor ligands such as
but represents an abnormal remodeling process resulting
transforming growth factor (TGF)-β1, wingless-type
in failure of the entire joint (5). At present, molecular
(Wnt), and Indian hedgehog (Ihh); signaling molecules
mechanisms of degeneration of articular cartilage, syno-
such as β-catenin and hypoxia-inducible factor (HIF)-
vial immunopathogenesis, and changes in subchondral
2α; and peptidases such as matrix metalloproteinase
bone are well accepted to be involved in the complex
(MMP) 13 and A Disintegrin and Metalloproteinase
initiation and progression of the disease.
with Thrombospondin Motifs (ADAMTS)-4/5.
As a unique component of the joints, articular car-
In the past, OA was regarded as a non-inflammatory
tilage has received much of the attention in OA studies
form of arthritis, but the truth is that synovitis is linked
because its damage is usually the most obvious patho-
to the increased cartilage damage (9). Recent studies
logic feature prior to joint dysfunction. The tissue
suggest that a large number of OA patients experience
structure can be divided into four zones: the superficial
pain consequent to synovitis (10), plus immune factor
zone, the transitional zone, the radial zone, and the

CONTACT Lunhao Bai bailh@sj-hopital.org Department of Orthopaedic Surgery, Sheng Jing-Hospital, China Medical University, SanHao Street #36,
HePing District, ShenYang, China. Tel: 18940251711.
© 2016 Taylor & Francis
246 Y. WEI AND L. BAI

(infiltrating macrophage, immunoglobulin, and I receptor can phosphorylate R-Smads at the


immune complex) detection in the synovial tissue of C-terminus of Smad2/3 (19). The phosphorylated
OA patient (7,11,12). Synovial inflammation may Smad2/3 then dissociates from the receptor complex
therefore be associated with OA. This review will and forms a heteromeric complex called Smad4,
focus on the impact of synovial inflammation on OA. which translocates into the nucleus and associates
We will discuss recent developments in (I) the correla- with other DNA-binding proteins to regulate gene tran-
tion between synovitis and OA, (II) innate immunity scription (20,21). The growth factor TGF-β, which
and the development of synovial inflammation (syno- strongly inhibits articular chondrocyte hypertrophy
vitis) (including cellular factors, the complement sys- and maturation, also represents a potential mechanism
tem, chemokines and cytokines), and (III) the role of in the development of OA.
meniscus, metabolic inflammation and nuclear factor Mice with the Smad3 gene knockout show chondro-
(NF)-κB in OA. cyte hypertrophy at an early stage, followed by
Growing evidence suggests that OA is an organ- decreased proteoglycan production, articular surface
level failure involving not only the cartilage surface fibrillation, and, in the late stage, chondrocyte cluster-
but also adjacent organizational structures. Changes ing in the deeper zone of articular cartilage, changes
in the subchondral bone can explain clinical symp- similar to those observed in human OA (21–23).
toms such as osteophyte formation. The importance Smurf2 is an inhibitor of TGF-β signaling in articular
of subchondral bone remodeling has been empha- chondrocytes and acts to promote chondrocyte hyper-
sized not only in humans but also in animal models trophy. Smurf2 is highly expressed in human OA car-
of acquired OA (13). Studies suggest that various tilage but is not present in normal cartilage. TGF-β
factors released from the subchondral bone environ- signaling is decreased, and expression of chondrocyte
ment are able to regulate cartilage metabolism (6,13) hypertrophic markers (ColX and MMP13) is increased
and could therefore become critical therapeutic tar- in transgenic mice (24), leading to progressive articular
get to control the disease, improving both clinical cartilage degradation, including reduced cartilage area
symptoms and joint structural erosion. This review and fibrillation, as well as subchondral sclerosis and
will summarize the current knowledge on the role of osteophyte formation (25). These findings suggest that
subchondral bone remodeling in the development of loss of TGF-β signaling represents one of the possible
OA through the induction of an abnormal pheno- mechanisms underlying OA development.
type in osteoblasts of subchondral bone and the
effects of chondrocytes on subchondral bone.
Effect of the Wnt/ß-catenin signaling pathway on
Degenerative change of articular chondrocytes articular chondrocyte degeneration in OA
in OA
As a key molecule in the canonical Wnt signaling
In recent times, there has been an explosion of reports pathway, β-catenin controls multiple critical pro-
describing abnormalities in the gross appearance, mate- cesses acting on the progression of OA (26). Wnt
rial properties, biochemical composition, cellular signaling can also promote cartilage degradation by
morphologies, and gene expression of articular carti- activation of catabolic genes like MMPs in chondro-
lages from human to animal models with OA-like cytes (27). Cytokine IL-1 was known as an activator
joints (14–16). These recent, detailed findings related of the WNT pathway involved in cartilage destruc-
to the molecular mechanisms of OA development will tion during OA (28), When Wnt (especially Wnt5
be summarized below (Figure 1b). and Wnt7) binds to its receptor Frizzled and the co-
receptors lipoprotein receptor-related protein (LRP)
5/6, the activity of the downstream signaling proteins
Effect of TGF-β in articular chondrocyte
Dishevelled (Dsh) and Axins 1 and 2 are altered. This
degeneration in OA
leads to the inactivation of Ser/Thr kinase glycogen
Intracellular chondrocyte signaling is initiated by TGF- synthase kinase (GSK)-3β, thus inhibiting the ubiqui-
β, through the combined TGF-β type I and II trans- tination and degradation of β-catenin triggered by
membrane Ser/Thr kinase receptors (17). TGF-β first GSK-3β. β-catenin then accumulates in the cytoplasm
binds to the type II receptor, triggering recruitment of and translocates into the nucleus, where it binds to
the type I receptor. This active type II receptor has the transcription factors lymphoid enhancer factor/T cell
ability to phosphorylate the GS domain of the type I factor (LEF-1/TCF) to regulate the transcription of
receptor (18). Gradually the accumulated activated type downstream target genes (29,30).
RECENT MOLECULAR MECHANISMS OF OSTEOARTHRITIS 247

Figure 1. Molecular mechanisms involved in the development of osteoarthritis including (a) complex cellular interplay in the
articular cavity (mainly synovial inflammation (synovitis) with OA). Synoviocytes (fibroblasts and macrophages) actively synthesize
proteases and cytokines—including IL-6, IL-8, and TNF-α among others—which can negatively affect the articular cartilage.
Degenerative meniscus results in an accelerated progression toward cartilage degradation through elevating inflammatory cytokines,
specifically IL-1 and TNF-α. Chondrocytes may also upregulate apoptosis and ECM degradation, resulting in diminished local
cellularity and eventually, cartilage loss. (b) Molecular mechanisms of articular chondrocyte degenerative changes in OA. The
diagram shows the involvement of receptor ligands such as TGF-β, Indian hedgehog, Wnt-β-catenin, NF-κB signal, and the HIF-2a
signaling pathway, which ultimately activate MMP-13 and ADAMTS, causing cartilage degeneration. (c) Molecular and cellular
interface between chondrocytes and subchondrocytes and osteoblasts at the osteochondral junction in OA. The presence of
connections (microfissures, vascular channels and diffusion) between subchondral bone and cartilage contributes to elevated
crosstalk between chondrocytes. Exposing subchondral bone to cytokines and growth factors from chondrocytes, such as VEGF,
promotes osteoclast activation and increases subchondral bone remodeling, leading to osteoblast abnormality and osteophyte
formation.

Appropriate levels of β-catenin may maintain Wnt-induced signaling protein 1 (WISP-1) in the
moderate maturation of articular chondrocytes mouse knee joint also leads to cartilage impairment.
under physiological conditions (31). Both excessive One study has shown that overexpression of β-catenin
and insufficient β-catenin levels may destroy the in articular cartilage causes abnormal differentiation of
homeostasis of articular chondrocytes by enhancing articular chondrocytes, resulting in cartilage degenera-
pathological maturation and apoptosis, respectively. tion (37).
The Wnt/β-catenin signaling pathway induces
MMP13 expression and the expression of aggrecan
Effect of Indian hedgehog on articular chondrocyte
through the anabolic factor TGFβ-3, leading to
degeneration in OA
degeneration and development of OA-like chondro-
cytes (32,33). Indian hedgehog (Ihh), as a member of the hedgehog
Recent findings implied that β-catenin was upregu- protein family, plays a crucial role in chondrocyte pro-
lated in articular cartilage tissue derived from patients liferation and differentiation (38). In mature articular
with OA (34). Moreover, protein secreted Frizzled- cartilage the Ihh signaling pathway also plays a critical
related protein (Sfrp)3, a secreted negative regulatory role during OA development. Ihh signaling acts
factor of Wnt signaling, and mutations of the Frizzled- through two transmembrane receptors: patched (Ptch)
related protein (FrzB) gene can increase susceptibility to and smoothened (Smo). In the absence of the Ihh
OA (35). FrzB knockout mice are susceptible to chemi- ligand, Ptch binds to Smo leading to inhibition of its
cal-induced OA (36). Furthermore, overexpression of function. Once Ihh signaling is activated, Ihh binds
248 Y. WEI AND L. BAI

Ptch leading to the release of Smo. Smo will further sensitive subunit HIF-1 severely interfered with proper
activate Gli transcription factors to regulate Ihh- skeletal development and led to massive cell death
responsive genes (39). within the center of the forming cartilaginous elements
A recent study reported that Ihh is upregulated in (49,50). In vitro studies show that HIF-2α is a potent
human OA cartilage and synovial fluid, and that the trans-activator of OA marker genes, including ColX,
elevated level correlates with OA progression (40). The MMP13, and VEGF (47). Overexpression of HIF-2α
abnormality observed in Ihh–/– mice is an increase in by intra-articular injection of adenovirus expressing
the fraction of chondrocytes which are post-mitotic, HIF-2α (Ad-EPAS1), the gene HIF-2α encodes, may
hypertrophic chondrocytes. This abnormality results lead to a spontaneous mouse model of OA (29).
from immature chondrocytes because Ihh–/– cartilage Moreover, heterozygous EPAS1-deficient mice have
fails to synthesize parathyroid hormone-related protein resistance to surgery-induced OA progression in a
(PTHrP) that keep chondrocytes in the proliferative mouse model (47,48). Some studies also suggest that
phase and correlate with chondrocyte phenotype, spe- NF-κB is the upstream inducer of HIF-2α expression
cifically decrease expression of SOX9 leading to and upregulates NF-κB signaling. HIF-2α binds to β-
increased COL2A1 (collagen type II, α1) expression catenin and enhances the transcriptional activity of β-
(41,42). Ihh is a critical and possible direct regulator catenin/T cell factor (TCF) by recruiting p300. The
of joint development. In its absence, distribution and actions of HIF-2α, interestingly, oppose those of HIF-
function of growth and differentiation factor (GDF)5- 1α on β-catenin, which suggests that the balance
expressing interzone-associated cells are abnormal (43). between HIF-1α and HIF-2β might play an important
A mouse genetic study further provided direct evidence role in the cell when hypoxia and Wnt stimulation
that conditional deletion of Ihh in chondrocytes attenu- coexist (51). These observations suggest that the HIF-
ates OA progression (44). These findings provide 1α/HIF-2α signaling pathway is also involved in OA
strong evidence that Ihh plays a critical role in OA development.
pathology, being a central regulator of OA progression,
and raise the possibility that blocking Ihh signaling
Effect of MMP13/Sox 9/ADAMTS on articular
could be used as a novel therapy to treat articular
chondrocyte degeneration in OA
cartilage degeneration in OA.
Chondrocytes constitute the unique cellular component
of articular cartilage. These cells synthesize the compo-
Effect of hypoxia-inducible factor-2α on articular
nents of the extracellular matrix (ECM), including col-
chondrocyte degeneration in OA
lagens, proteoglycans, and non-collagenous proteins. In
The HIF proteins belong to a transcription factor OA, cartilage homeostasis and synthesis of the ECM are
family, and consist of α and β subunit members. HIF- disrupted by many biophysical and biochemical factors.
1 and HIF-2 are key effectors of the cellular response to In this review, the mechanisms described may only
hypoxia, and their activity is directly regulated by the provide insights into the roles of the core factors—
level of oxygen (45,46). Under normoxic conditions, MMP13, ADAMTS, SOX9—in the pathogenesis of OA.
the proline residues on the α-subunits are hydroxylated; Human clinical and animal studies show that
these are recognized by the von Hippel–Lindau protein MMP13 plays a dominant role in the progression of
(pVHL) tumor suppressor, an E3 ubiquitin ligase, and cartilage degeneration (52). MMP13 is a collagenase
degraded by proteasomes. In contrast, under hypoxic with substrate specificity that targets collagen for degra-
conditions, HIF-α is not hydroxylated, enabling it to dation. Compared to other MMPs, MMP13 has a more
heterodimerize with β-subunits known as the aryl restricted expression pattern in connective tissue (53).
hydrocarbon receptor nuclear translocator (ARNT), MMP13 has a much higher catalytic rate than other
ARNT2, ARNT-like (ARNTL), and ARNTL2 to activate MMPs for ColII and gelatin, which makes it the most
target genes (45,47). potent peptidolytic enzyme among collagenases (29).
Studies show that the expression levels of HIF-2α are MMP13 preferentially cleaves ColII, which is the most
significantly increased in both human and mouse OA abundant protein in articular cartilage. Cleavage of the
cartilages (47,48). Due to the lack of vascular tissue in Gly978–Gln979 bond occurs at a specific location (at ¾
articular cartilage, chondrocytes in the deeper zones length site of type II collagen) (54). MMP-13-overex-
exist under conditions of low oxygen tension. The pressing transgenic mice developed spontaneous articu-
importance of HIF-1 in the formation and maintenance lar cartilage destruction characterized by excessive
of cartilage tissue has been demonstrated in conditional cleavage of Collagen II and loss of aggrecan (55).
knockout mice in which deletion of the oxygen- There is considerable evidence that proteolysis of
RECENT MOLECULAR MECHANISMS OF OSTEOARTHRITIS 249

aggrecan by ADAMTS occurs before and indeed may immune system and inflammatory response in the
be prerequisite for MMP-driven collagenolysis. It is also pathogenesis of the disease. Despite a long history of
apparent that increased cleavage of collagen by MMP- categorizing OA as a non-inflammatory form of arthri-
13. Type II collagen can restrict the swelling pressure of tis, pathologic studies of specimens in the 1980s
aggrecan and its loss also lead to the release of aggrecan described synovial inflammatory infiltrates of mono-
and loss of normal viscoelastic function of cartilage nuclear cells (Figure 1a) (66).
(56). Moreover, the expression of MMP-13 is signifi-
cantly increased in articular cartilage in β-catenin-con-
Correlation between synovitis and OA
ditional activation mice (57). These findings implicate
MMP-13 as a critical player in the progression of OA Despite the different approach employed in the detec-
that could serve as a molecular target for OA therapies. tion and characterization of synovitis (such as imaging
Sox9 is critical for chondrocyte differentiation and or histologic assessment) (67–69), published studies
cartilage morphogenesis during skeletal development consistently provide evidence of a correlation between
(58). A gene expression study reported that Sox9 synovial inflammation and symptoms such as pain
expression was lower in human OA cartilage (59). (linked to inflammation) in patients with knee OA
Sox9, a key chondrogenic transcription factor, plays a (70,71). Scanzello et al. have contributed further evi-
crucial role in the development and maintenance of the dence of an association between synovitis (defined his-
chondrogenic phenotype. Saito T found that the over- tologically) and knee symptoms measured by the
expression of SOX-9 in vitro can mediate the inhibition Lysholm score (which measures pain, swelling, limping,
of terminal differentiation of chondrocytes and keep it locking, instability, and functional disability on a single
“fresh” (60). Therefore, modulation of OA cartilage by scale) in patients with early knee OA undergoing
genetically modifying the levels of Sox9 expression may arthroscopic meniscectomy (72). Synovitis has been
be advantageous in ameliorating the course of OA (61). related not only to knee pain but also to knee joint
ADAMTS are another family of enzymes targeting function, using objective outcome measures of walking
collagens and aggrecan for proteolysis, and are major and stair-climbing times (72,73). These efforts have
contributors to cartilage degeneration in OA. Studies provided substantial evidence that synovitis is asso-
show that there are two types of aggrecanase: ciated with major symptoms such as pain and degree
ADAMTS4 and ADAMTS5 (62). Expression and activity of joint dysfunction, and may promote more rapid
of ADAMTS4 have been shown to be upregulated during cartilage degeneration (69,74).
cartilage degeneration, while ADAMTS5 has been shown
to be active in both normal and degenerated cartilages
Innate immunity of synovial inflammation
(63,64). Surgically induced OA experiments performed in
(synovitis) in OA (Table 1)
both ADAMTS4 and ADAMTS5 knockout mice showed
that deletion of the ADAMTS5 gene alone relieved carti- Monocytes/macrophages and other cells of the
lage degradation, while deletion of the ADAMTS4 gene innate immune system in OA
could not (64), suggesting that ADAMTS5 may play a Macrophages are among the most abundant cell types
more important role in OA development than present in cellular infiltrates found in the inflamed
ADAMTS4. In fact, study of ADAMTS5 knockout mice synovium of OA patients (75,76). Monocyte subsets,
showed that the protection was due to elimination of classic, intermediate, and nonclassic, play a major
fibrous overgrowth from the periarticular tissues and homeostatic role in tissue remodeling and mainte-
deposition of newly synthesized aggrecan in the cartilage nance, facilitated by apoptotic “eat me” opsonins like
(65). The effect of ADAMTS5 on collagenous tissues may C-reactive protein (CRP), serum amyloid P, C1q, C3b,
be revealed by an analysis of Smad-signaling pathways in IgM, ficolin, and surfactant proteins (77). Monocyte
wild-type and ADAMTS5-deficient fibroblasts and chon- chemoattractant protein 1 (CCL2 or MCP-1) is one of
drocytes. These animal model studies will definitely influ- the most important chemokines regulating systemic
ence strategies employed in the therapeutic control of and local cell trafficking and infiltration of monocyte/
human OA in the future. macrophages in chronic inflammation. Blocking CCL2-
CCR2/CCR4 ligand–receptor axis using 7ND, a mutant
CCL2 protein, can reduce migration of monocytes/
Inflammatory response and
macrophages in vitro (78). Using a collagenase-induced
immunopathogenesis
mouse model, Bondeson et al. found several chemo-
Since OA is considered a whole joint disease, increasing kines responsible for chemotaxis of macrophages in the
evidence has demonstrated the involvement of innate development of OA, such as TGF-β and interleukin
250 Y. WEI AND L. BAI

Table 1. Molecular factors of inflammatory response and immunopathogenesis of Osteoarthritis (OA).


Expression represents duration of synovial
Molecular factors Function in OA procession inflammation in OA References
TGF-β responsible for chemotaxis and activation of macrophages Innate immunity of Synovial Inflammation in (75)
IL-1β OA (76)
IL-6 (77)
IL-8
MMP-1 (77)
MMP-3
TLRs a group of motif recognition receptors important in eliciting (85–87)
an initial innate response
decay accelerating factor activation of complement-mediated response Activation of complement system of Synovial (92)
(DAF) Inflammation in OA
C5b-9 terminal complement (93)
complex
fibromodulin positive affect complement cascade activity (100)
cartilage oligomeric matrix (101)
protein (COMP)
osteoadherin (102)
collagen IX inhibitors of complement cascade activity (103)
MAC complex maybe a protection in development of OA (104)
IL-1 relive the clinical symptom Cytokines of Synovial Inflammation in OA (108)
NALP3 inflammasome et al. Responsible for IL-1 activation (109)
TNF-α participates in chondrocyte destruction and pain symptom (112,113)
IL-6 pro-inflammatory cytokines in the development of OA (104)
IL-7
IL-8
leukemia inhibitory factor
(LIF)
IL-11
IL-17
IL-18
oncostatin M(OSM)
IL-4, anti-inflammatory cytokines in the development of OA
IL-10
IL-13
IL-8/CXCL-8 induce hypertrophic differentiation and calcification in Chemokines of Synovial Inflammation in OA (63)
GRO-α/CXCL-1 chondrocytes
RANTES/CCL-5 and its induced normal chondrocytes collageⅡdegradation
receptors
CCR7 a role in synovial angiogenesis when OA starting (67)
CCL19
MIP-1γ/CCL-9 increased formation of osteoclasts in joints (115)

(IL)-1β (79,80). Macrophage depletion also resulted in activation in the SM is an important stimulus for
decreased production of IL-6, IL-8, MMP-1, and MMP- recruiting and activating macrophages, granulocytes
3 (81). These findings suggest that activation of syno- and lymphocytes (92). Evidence has suggested that a
vial macrophage is required for the production of number of endogenous products, produced by matrix
MMPs leading to cartilage damage (82–85). Natural disruption or cellular stress, can also bind and activate
killer cells have also been obtained from synovial tis- TLRs. Many putative TLR ligands are modified in form
sues of patients undergoing total joint replacements, or distribution, or increased in concentration in joint
but the mechanism of pathogenesis has not yet been fluids or tissues in the setting of OA joint (93). The
elucidated in detail (86). regulatory processes involved in TLR activation are
Activation of the innate response often begins with complex, and their role in promoting synovitis in OA
the stimulation of pattern-recognition receptors is not fully established. However, targeting TLRs,
(PRRs), classically in the setting of infectious insult by ligands, and pathways that trigger their activation
microbial ligands (87). However, Under OA progres- need to be explored as potential therapeutic approaches
sion, PRRs can be activated by endogenous damage- in OA.
associated molecular patterns (DAMPS) such as hyalur-
onan, rather than by microbial ligands (88). TLRs
Activation of the complement system in synovial
(TLR1 through 10 in humans) are a group of motif
inflammation (synovitis) in OA
recognition receptors important in eliciting an initial
The complement system, including a cascade of pro-
innate response against pathogens. TLRs are constitu-
teins, constitutes an important effector mechanism in
tively expressed on macrophages mostly (89). TLRs 1–7
the immune system to eliminate pathogens and
and 9 have been detected in SM in OA (90,91). TLR
immune complexes. The deposition and activation of
RECENT MOLECULAR MECHANISMS OF OSTEOARTHRITIS 251

complement factors in OA cartilage have been docu- receptor antagonist, which blocks IL-1 activity, is produced
mented in several studies in OA patients as well as in by synoviocytes at higher levels in OA (98). Production of
animal models of OA (94,95). Tarkowski et al. observed active IL-1 requires activation of the NALP3 inflamma-
expression of decay accelerating factor (DAF) in the some by agonists such as crystals including basic calcium
synovial lining cell layer in OA along with C5b-9 term- phosphate (113) and hydroxyapatite (114). Crystal
inal complement complex suggesting activation of a Deposition of both hydroxyapatite and basic calcium phos-
complement-mediated response (96,97). However, phate occurs in patients with OA. Therefore, it is predicted
blood or serum leaking into the joint after injury pro- that IL-1 may be an important effector of crystalline and
vide a source of complement proteins in many patients other crystals’ (basic calcium phosphate and hydroxyapa-
(98–101), and as early as 1988, Corvetta et al. found tite) deposition in patients with OA.
complement deposition in synovial membranes follow-
ing meniscal tears or cartilage degeneration (102). Tumor Necrosis Factor (TNF)-α. TNF-α was identified
Chondrocytes and synovial macrophages may also in the SF of patients with OA as early as in 1996.
actively produce complement components and inhibi- Although TNF-α is elevated in OA, it is not as high
tors (97,103). Complement components and immuno- as that seen in RA. (115). TNF also participates in
globulins have been identified in synovial fluid from chondrocyte destruction (116). Evidence suggested
OA patients using proteomic approaches (98). Several that the synovium supernatants TNF-α concentrations
studies have also demonstrated that other molecular were significantly higher in supernatants from cultured
components in the articular cartilage may affect com- OA synovium as compared with non-arthritic (NA)
plement cascade activity, such as fibromodulin (104), synovium (117). TNF receptor (TNF-R) becomes upre-
cartilage oligomeric matrix protein (COMP) (105), or gulated on chondrocytes in cartilage from specific
osteoadherin (106). In contrast, collagen IX, properdin, regions (118). The answer may lie in the differences
and CD59 can act as inhibitors of complement (107). In in weight bearing experienced by chondrocytes in car-
recent studies, mice with impaired ability to generate tilage from different regions of the joint. Loading has a
the complement membrane attack complex (MAC) profound effect on chondrocyte metabolism. Magnano
were partially spared from the development of OA, et al. reported the results of a trial of a TNF-inhibitor in
showing direct evidence of role of complement system an open-label pilot study to treat pain and inflamma-
in OA pathogenesis (108). However, the way comple- tion in twelve patients with OA (119). Like the IL-1
ment deposition occur in joint synovium of OA, and trials, this trial did not demonstrate apparent improve-
the role of the complement cascade in the pathogenesis ment, but nevertheless improvements in pain and phy-
remain to be precisely determined. sical function scores were reported by some individuals.
This may reflect a more complex interplay involving
Role of cytokines in the synovial inflammation of OA various pathogenic factors in OA.
PPRs and activation of the complement cascade result in
the release of several soluble factors such as cytokines (IL- Other cytokines. The study of the roles of other pro-
1, TNF-α, etc.) and chemokines (IL-8, CCL5, CCL19, and inflammatory cytokines, including IL-6, oncostatin M
its receptor CCR7) from the synovium causing further (OSM), IL-7, IL-8, leukemia inhibitory factor (LIF), IL-
progression of OA symptoms (109). During OA, these 11, IL-17, IL-18, and anti-inflammatory cytokines
mediators may be produced by a variety of cell types, including IL-4, IL-10 and IL-13, show that synovial
including macrophages, chondrocytes, and synovial fibro- inflammation impacts on chondrocyte metabolism,
blasts (110). Such cytokines as synovial factors have and hence, favoring the development of OA (110).
shown much potential to induce cartilage destruction in
vitro (111). Some specific examples will be introduced Chemokines. Chemokines are small secreted mole-
below with respect to synovial inflammation in OA. cules responsible for chemotaxis of immune cells
and thus play an important role in the immune sys-
IL-1. Although IL-1 has significant effects on articular tem. Scanzello demonstrated that synovial inflamma-
cartilage destruction, IL-1 is not consistently elevated in tory infiltrates were associated with expression of a
the synovial fluid of OA patients (10). Attempts to block distinct mRNA chemokine in patients with OA. The
IL-1 activity therapeutically in patients have been asso- chemokine included IL-8, CCL5, CCL19, and its
ciated with relief of the clinical symptom until 3 months receptor CCR7 (72). Merz et al. showed that IL-8/
(112); articular cartilage destruction was not improved. CXCL-8 may induce hypertrophic differentiation and
However, it is possible that IL-1 activity may be important calcification of chondrocytes (120). While CCR7 is
in specific clinical settings of OA. The endogenous IL-1 expressed by synovial fibroblasts, and, when activated
252 Y. WEI AND L. BAI

by its ligand CCL19, mediates upregulation of vascu- reduced function. The addition of elevated inflamma-
lar endothelial growth factor (VEGF), implying a role tion at the time of meniscal damage or meniscal degen-
in synovial angiogenesis in the early stages of OA eration results in an accelerated progression toward
(79). CCR5 together with its ligand CCL-21 plays a cartilage degradation. Shortly after a meniscus rupture,
similar role to CCL19. MIP-1γ/CCL-9 has been inflammatory cytokines, specifically IL-1 and TNF-α,
shown to be produced by activated CD4+ T cells are upregulated in the joint, indicating an inflammatory
present in the synovium of a mouse model of OA, response (129). Specifically, Scanzello et al. also found
which also resulted in increased formation of osteo- that synovial IL-15 is elevated in degenerative meniscal
clasts in the joints (121). In summary, chemokines tear patients with Kellgren–Lawrence grade 1 or 2
represent a class of soluble inflammatory mediators compared to end-stage OA patients undergoing total
that have pleiotropic effects on joint tissues, and may knee replacement (130). Hellio Le Graverand et al. used
contribute to inflammation and clinical symptoms in ACL-transected rabbits as an OA initiation model in
patients with OA. order to measure inflammatory expression changes in
the early stages during which the meniscus is degener-
ating. This study found that while meniscal expression
Role of inflammatory signal NF-κB in OA
of TNF-α was downregulated, the inflammatory
The abundant form of NF-κB is a heterodimer of p65/ enzyme COX-2 (cyclooxygenase-2) was upregulated in
RelA and p50. In the resting state, this is bound to the the medial meniscus 3 weeks after ACL transaction,
inhibitory protein IκB of which αorβ are the major indicating an inflammatory response in the degenerat-
forms. The trimeric complex in mainly cytoplasm. ing meniscus during the early stages of OA develop-
When a signaling complex forms, it activates the κB ment. Thus, elevated levels of inflammation may be
kinase (IKK) component of the IKK complex. The IKK present during meniscal degeneration (131). Meniscal
complex is also a trimer and comprises IKKα and β, cells are also responsive to cytokine stimulation like IL-
together with IKKγ. IKKγ is an important adaptor 1β, IL-6, and TGF-α with increased proteoglycan and
enabling the receptor signaling complexes to activate nitric oxide release (132). The degenerated menisci
the IKKβ. IKKβ phosphorylates IκBα at Ser32 and show significant upregulated expression of ADAMTS
Ser36. The phosphorylations target the κB for rapid and MMPs following cytokine stimulation, which con-
ubiquitination and degradation. This removes the tributed to cartilage degeneration (133). These pieces of
nuclear export signal of the inhibitor and unmasks a evidence suggest that the addition of elevated inflam-
nuclear localization signal on the p65 subunit. The NF- mation at the time of meniscal damage or meniscal
κB dimer therefore locates mainly in the nucleus where degeneration results in an accelerated progression
it binds to specific sites in the promoter regions of toward cartilage degradation. Cell adhesion markers
sensitive genes and promotes transcription (122). NF- such as vascular cell adhesion molecule (VCAM)-1,
κB transcription factors can be triggered by a host of intercellular adhesion molecule (ICAM)-1, and
stress-related stimuli like pro-inflammatory cytokines E-selectin were also increased in the degenerative
IL-1 (123); indeed, through the canonical NF-κB path- menisci and were to be present in hypertrophic and
way to trigger the expression of metalloproteinases early osteoarthritic synoviumn and were involved in
(MMPs), prostaglandin E2 (PGE2), nitric oxide (NO), inflammatory cell recruitment to the synovium
and type X collagen in chondrocytes (124,125), leading (134,135).
to increased cartilage degradation and chondrocyte
hypertrophy in OA cartilage.
Pathogenic role of metabolic triggered
inflammation in OA
Role of meniscus in the synovial inflammation of
The incidence of obesity worldwide has increased dra-
OA
matically in recent decades. Accordingly, obesity and
The clinical importance of the meniscus in OA devel- associated disorders such as OA now constitute a ser-
opment is well documented (126–128). However, ious threat to the current and future health of both
meniscus pathology in OA is largely attributed to developed and developing populations. A newly
mechanically mediated loss of structural integrity. defined phenotype of OA, “metabolic OA,” has been
Accumulating evidence supports that knee OA progres- associated with metabolic syndrome (MetS) and obesity
sion is often driven by biomechanical forces, and the (136). Metabolic triggered inflammation also called
pathological response of tissues to such forces leads to meta-inflammation (137) is mainly caused by nutrient
structural joint deterioration, knee symptoms, and overload and metabolic surplus (138). Metabolic
RECENT MOLECULAR MECHANISMS OF OSTEOARTHRITIS 253

overload results in oxidative stress and inflammation, Subchondral osteoblasts have an abnormal
which then triggers vicious stress cycles that lead to cell phenotype in OA
dysfunction (139). The components that make up the
Subchondral bone sclerosis is characterized by trabecu-
cluster of MetS, such as overweight, dyslipidemia, and
lar thickening, an increase in osteoid volume, and a
impaired glucose tolerance, have been involved in
decrease in calcium binding to collagen fibers. This
meta-inflammation (140). Meta-inflammation usually
abnormal mineralization is due to the overproduction
is a type of chronic inflammation presenting elevated
of the homotrimeric α1 form of type I collagen by
levels of cytokines, acute-phase inflammatory compo-
osteoblasts. This (α1)3 type I collagen has lower affinity
nents (complements and CRP), and other mediators
for calcium than (α1)2 type I collagen (152,153).
(141,142). Furthermore, adipocytes, as the key cell of
Increasing evidence suggests that OA osteoblasts
meta-inflammation, also synthesize numerous cyto-
showed a decreased cAMP response to PTH and an
kines such as IL-6, IL-1, and TNF-α and adipokines
increased production of alkaline phosphatase and
such as leptin, adiponectin, resistin, and visfatin (143).
osteocalcin in response to vitamin D3. Further, under
Excessively secreted factors could disturb the integra-
basal conditions, OA osteoblasts produce more insulin-
tion of systemic metabolism, thereby leading to inflam-
like growth factor-1 and urokinase than normal cells. It
matory response. In addition, some local fat tissues,
has also been reported that subchondral bone explants
such as infra-patellar fat pad (IPFP), may act as mod-
from OA patients produce more IL-6, IL-8, and MMP-
ulators in OA. IPFP was considered a source of local
13 (154) as well as osteopontin, osteocalcin, TGF-β1,
inflammatory mediators and thus an active osteoar-
type I collagen, IL-6, IL-8, and high levels of alkaline
thritic tissue (144), but it also has a beneficial effect
phosphatase (ALP) but downregulated osteoprotegerin
possibly through biomechanical mechanisms, as sup-
(OPG) gene expression and protein production (155–
ported by a recent study suggesting that IPFP size was
157). IL-6 and PGE2 stimulate receptor activator of
negatively associated with OA disease severity indepen-
nuclear factor Kappa-B-/Ligand (RANKL) and inhibit
dent of BMI and total body fat (145).
OPG production by osteoblasts and thus increase
osteoclast differentiation and activation via their effect
Subchondral bone changes in OA on the RANK/RANKL/OPG system (158,159). These
factors play a dominant role in osteoclastic activity.
Subchondral bone plays a critical role in OA progres-
Therefore, the bone-forming activity of subchondral
sion. The term subchondral bone refers to a layer of
bone cannot be accompanied by equivalent mineraliza-
bone immediately beneath the cartilage and is formed
tion. In addition, osteoblast showed an increased
by a 6-mm layer of trabecular bone plate. Anatomically,
expression of MMP-1which would account for degra-
the subchondral bone consists of both a subchondral
dation of non-mineralized collagen type I in SBC (160).
cortical plate and a layer of cancellous bone. The sub-
These studies suggest that the formation of unminer-
chondral cortical plate is in nature similar to other
alized, immature new bone leads to the appearance of
cortical bone, while subchondral cancellous bone has
abundant osteoids and bone cysts in OA.
a lower volume, density, and stiffness than the cortical
plate (146). However, based on the available evidence,
there is no consensus on subchondral bone changes in
Cartilage interface with subchondral bone in OA
early OA. Some studies have previously suggested that a
thickening of the growth plate is occurring as a primary In OA, chondrocytes are stimulated to express molecules
fundamental subchondral bone change in OA (147). It that are associated with chondrocyte hypertrophy and
has also been demonstrated in animal studies that thin- terminal differentiation, such as VEGF, runt-related tran-
ning of the subchondral bone occurred before cartilage scription factor 2 (RUNX2) and MMP-13. Secretion of
damage was detected, suggesting that bone turnover angiogenic factors such as VEGF increases vascularity
may be increased in the early stages of OA (148). within the deep layers of articular cartilage, thus facilitat-
Surely, changes to subchondral bone in the progressive ing molecular transport. Recently, the communication
stages of OA are characterized by extensive remodeling pathways of cartilage effect on bone reported includes
of the trabeculae, formation of new bone below new microcracks, neovascularization, vascular channels next
cartilage (osteophytes), and the development of sub- to the cruciate ligaments, in addition, small molecules
chondral bone cysts (SBC) (149–151). Like cartilage, diffusion directly (161,162). Chemokines, cytokines, and
an increase in expression level of certain genes in proteases secreted by chondrocytes have been implicated
bone is regarded as a biochemical marker of OA in the alteration of biochemical and functional abilities of
(Figure 1c). the osteoblasts within subchondral bone. For example, IL-
254 Y. WEI AND L. BAI

6 in combination with other cytokines such as IL-1β can It is true that in past decades, increased knowledge of
switch osteoblasts from a normal phenotype to a sclerotic the detailed molecular and signaling pathways involved
phenotype (162,163). There is increasing evidence that in OA development has enabled significant progress to
the Wnt signaling pathway is a key regulator of subchon- be made which has even been applied to clinical treat-
dral bone and is implicated in OA (164). In an animal ment. However, what is the exact pattern of degradation?
model of OA, Wnt activation was predominantly What tissue is the lead problem in the early stage of OA?
observed in osteocytes of the subchondral bone. Altered How do tissues respond in later stages? More exact pre-
activity of Wnt signaling in chondrocytes may involve cision mechanisms need to be further explored. In addi-
osteoclastic activity in subchondral bone growth plates tion, in the clinic, many patients suffering similar degrees
leading to sclerosis or osteophyte formation at the edges of OA received the same drug and physical treatments,
of joints (165,166). In addition, Dickkopf-1 (Dkk1) is an but surprisingly, different outcomes were observed. This
inhibitor of this pathway, and its overexpression in bone shows that there are differences in OA-related genes
leads to thicker and stiffer bones, retarding cartilage between individuals. Recent advances in epigenetic stu-
degradation (167). Moreover, the deletion of sclerostin, dies have shed light on the importance of epigenetic
another Wnt antagonist, increased the severity of OA, regulation of gene expression in the development of
although no changes in cartilage were observed (168). OA. Microarray-based gene expression studies and net-
Dkk2 is regulated by vitamin D and TGFβ expression work-based analysis, along with epigenetic analysis, are
was increased and both contributed to the abnormal essential for the identification of signaling pathways
osteoblast phenotype in OA, which implied that TGFβ is regulated during disease development. The epigenetic
another key factor involved in development of bone findings may not only broaden our understanding of
abnormalities during OA, promoting both osteophyte the molecular mechanisms underlying the development
growth and subchondral bone formation (169,170). of OA, but also promote the development of new drugs
Lastly, numerous factors in the subchondral bone envir- for the treatment of OA. In the future, individualized and
onment have been identified as pathogenic factors affect- comprehensive treatment will be applied to the interven-
ing cartilage and could therefore be potential therapeutic tion of OA.
targets in OA.

Financial support
Conclusions and perspectives This work is supported by National Natural Science
Foundation of China (General Program; Grant
Collective evidence indicates that OA is not simply a
Number: 81272050).
process of wear and tear but a disease with more com-
plex etiology, more risk factors and more molecular
mechanisms resulting in organ failure of the entire Declaration of interest
joint. Among the molecular mechanisms, degeneration The authors declare no competing interests.
of articular cartilage, synovial immunopathogenesis,
and changes in subchondral bone are key factors in
the mechanisms leading to joint damage in OA. Thus, References
synovial change, cartilage and subchondral bone para-
1. Thomas E, Peat G, Croft P. Defining and mapping the
meters assessed by imaging techniques, or biochemical person with osteoarthritis for population studies and
markers may soon help the clinician to identify differ- public health. Rheumatology (Oxford, England)
ent specific OA phenotypes. This stratification strategy 2014;53(2):338–45.
is likely to expand the options for clinicians to use 2. Rannou F, Bertin P, Grange L, Branchoux S,
comprehensive methods to evaluate and treat OA Dachicourt JN, Taieb C. The burden of osteoarthritis:
development and validation of a new assessment tool
patients. Novel drugs and agents that specifically
(BONe’S). Curr Med Res Opin 2014;30(4):741–51.
block the mechanisms involved in synovial inflamma- 3. Silverwood V, Blagojevic-Bucknall M, Jinks C, Jordan
tion can potentially protect against degeneration of JL, Protheroe J, Jordan KP. Current evidence on risk
articular cartilage and prevent subchondral bone remo- factors for knee osteoarthritis in older adults: a sys-
deling responsible for the osteoarthritic changes, tematic review and meta-analysis. Osteoarthritis
thereby enabling targeted therapy to be developed. Cartilage 2015;23(4):507–15.
4. Lane NE, Brandt K, Hawker G, Peeva E, Schreyer E,
These comprehensive agents will benefit suffering Tsuji W, Hochberg MC. OARSI-FDA initiative: defin-
patients and provide exciting new hope for the treat- ing the disease state of osteoarthritis. Osteoarthritis
ment of OA. Cartilage 2011;19(5):478–82.
RECENT MOLECULAR MECHANISMS OF OSTEOARTHRITIS 255

5. Loeser RF, Goldring SR, Scanzello CR, Goldring MB. effects of TGF-beta on Smad2/3 Activity, cell migration
Osteoarthritis: a disease of the joint as an organ. and epithelial to mesenchymal transition. Mol
Arthritis Rheum 2012;64(6):1697–707. Pharmacol 2015;88(6):1062–71.
6. Bertrand J, Cromme C, Umlauf D, Frank S, Pap T. 20. Kaminska B, Kocyk M, Kijewska M. TGF beta signal-
Molecular mechanisms of cartilage remodelling in osteoar- ing and its role in glioma pathogenesis. Adv Exp Med
thritis. Int J Biochem Cell Biol 2010;42(10):1594–601. Biol 2013;986:171–87.
7. Ponchel F, Burska AN, Hensor EM, Raja R, Campbell 21. Piersma B, Bank RA, Boersema M. Signaling in fibro-
M, Emery P, Conaghan PG. Changes in peripheral sis: TGF-beta, WNT, and YAP/TAZ converge. Front
blood immune cell composition in osteoarthritis. Med 2015;2:59.
Osteoarthritis Cartilage 2015;23(11):1870–8. 22. Reynard LN, Loughlin J. The genetics and functional
8. Sherwood JC, Bertrand J, Eldridge SE, Dell’Accio F. analysis of primary osteoarthritis susceptibility. Expert
Cellular and molecular mechanisms of cartilage Rev Mol Med 2013;15:e2.
damage and repair. Drug Discovery Today 2014;19 23. Lee AM, Shimizu C, Oharaseki T, Takahashi K, Daniels
(8):1172–77. LB, Kahn A, Adamson R, Dembitsky W, Gordon JB,
9. Felson DT, Niu J, Neogi T, Goggins J, Nevitt MC, Burns JC. Role of TGF-beta signaling in remodeling of
Roemer F, Torner J, Lewis CE, Guermazi A. Synovitis noncoronary artery aneurysms in Kawasaki disease.
and the risk of knee osteoarthritis: the MOST Study. Pediatr Dev Pathol2015;18(4):310–17.
Osteoarthritis Cartilage 2016;24(3):458–64. 24. Holz JD, Beier E, Sheu TJ, Ubayawardena R, Wang M,
10. Scanzello CR, Goldring SR. The role of synovitis in Sampson ER, Rosier RN, Zuscik M, Puzas JE. Lead
osteoarthritis pathogenesis. Bone 2012;51(2):249–57. induces an osteoarthritis-like phenotype in articular
11. Wang Q, Rozelle AL, Lepus CM, Scanzello CR, Song JJ, chondrocytes through disruption of TGF-beta signal-
Larsen DM, Crish JF, Bebek G, Ritter SY, Lindstrom ing. J Orthopaedic Res2012;30(11):1760–66.
TM, Hwang I, Wong HH, Punzi L, Encarnacion A, 25. Chen CG, Thuillier D, Chin EN, Alliston T.
Shamloo M, Goodman SB, Wyss-Coray T, Goldring Chondrocyte-intrinsic Smad3 represses Runx2-induci-
SR, Banda NK, Thurman JM, Gobezie R, Crow MK, ble matrix metalloproteinase 13 expression to maintain
Holers VM, Lee DM, Robinson WH. Identification of a articular cartilage and prevent osteoarthritis. Arthritis
central role for complement in osteoarthritis. Nat Med Rheum 2012;64(10):3278–89.
2011;17(12):1674–79. 26. Wu Q, Zhu M, Rosier RN, Zuscik MJ, O’Keefe RJ,
12. Lee AS, Ellman MB, Yan D, Kroin JS, Cole BJ, van Chen D. Beta-catenin, cartilage, and osteoarthritis.
Wijnen AJ, Im HJ. A current review of molecular Ann NY Acad Sci 2010;1192:344–50.
mechanisms regarding osteoarthritis and pain. Gene 27. Yuasa T, Otani T, Koike T, Iwamoto M, Enomoto-
2013;527(2):440–7. Iwamoto M. Wnt/beta-catenin signaling stimulates
13. Karsdal MA, Bay-Jensen AC, Lories RJ, Abramson S, matrix catabolic genes and activity in articular chon-
Spector T, Pastoureau P, Christiansen C, Attur M, drocytes: its possible role in joint degeneration. Lab
Henriksen K, Goldring SR, Kraus V. The coupling of Invest 2008;88(3):264–74.
bone and cartilage turnover in osteoarthritis: opportu- 28. Ryu JH, Chun JS. Opposing roles of WNT-5A and
nities for bone antiresorptives and anabolics as WNT-11 in interleukin-1beta regulation of type II col-
potential treatments? Ann Rheumatic Dis 2014;73 lagen expression in articular chondrocytes. J Biol Chem
(2):336–48. 2006;281(31):22039–47.
14. Bijlsma JW, Berenbaum F, Lafeber FP. Osteoarthritis: 29. Wang M, Shen J, Jin H, Im HJ, Sandy J, Chen D.
an update with relevance for clinical practice. Lancet Recent progress in understanding molecular mechan-
(London, England) 2011;377(9783):2115–26. isms of cartilage degeneration during osteoarthritis.
15. Roemer FW, Guermazi A. Osteoarthritis year in review Ann New York Acad Sci 2011;1240:61–9.
2014: imaging. Osteoarthritis Cartilage 2014;22 30. Gough NR. Focus issue: Wnt and beta-catenin signal-
(12):2003–12. ing in development and disease. Sci Signaling 2012;5
16. Schroeppel JP, Crist JD, Anderson HC, Wang J. (206):eg2.
Molecular regulation of articular chondrocyte function 31. Lui JC, Chau M, Chen W, Cheung CS, Hanson J,
and its significance in osteoarthritis. Histol Histopathol Rodriguez-Canales J, Nilsson O, Baron J. Spatial regula-
2011;26(3):377–94. tion of gene expression during growth of articular car-
17. Huang SS, Chen CL, Huang FW, Johnson FE, Huang tilage in juvenile mice. Pediatr Res 2015;77(3):406–15.
JS. Ethanol enhances TGF-beta activity by recruiting 32. Zhang X, Zhu J, Li Y, Lin T, Siclari VA, Chandra A,
TGF-beta receptors from intracellular vesicles/lipid Candela EM, Koyama E, Enomoto-Iwamoto M, Qin L.
rafts/caveolae to non-lipid raft microdomains. J Cell Epidermal growth factor receptor (EGFR) signaling
Biochem 2016;117(4):860–71. regulates epiphyseal cartilage development through
18. Zuniga JE, Groppe JC, Cui Y, Hinck CS, Contreras- beta-catenin-dependent and -independent pathways. J
Shannon V, Pakhomova ON, Yang J, Tang Y, Mendoza Biol Chem 2013;288(45):32229–40.
V, Lopez-Casillas F, et al. Assembly of TbetaRI: 33. Naito M, Ohashi A, Takahashi T. Dexamethasone inhi-
TbetaRII:TGFbeta ternary complex in vitro with recep- bits chondrocyte differentiation by suppression of
tor extracellular domains is cooperative and isoform- Wnt/beta-catenin signaling in the chondrogenic cell
dependent. J Mol Biol 2005;354(5):1052–68. line ATDC5. Histochem Cell Biol 2015;144(3):261–72.
19. Lin H, Li N, He H, Ying Y, Sunkara S, Luo L, Lv N, 34. Shin Y, Huh YH, Kim K, Kim S, Park KH, Koh JT,
Huang D, Luo Z. AMPK inhibits the stimulatory Chun JS, Ryu JH. Low-density lipoprotein receptor-
256 Y. WEI AND L. BAI

related protein 5 governs Wnt-mediated osteoarthritic transcription factors, and renal cancer. Eur Urol
cartilage destruction. Arthritis Res Ther 2014;16(1): 2016;69(4):646–57.
R37. 47. Saito T, Fukai A, Mabuchi A, Ikeda T, Yano F, Ohba S,
35. Baker-Lepain JC, Lynch JA, Parimi N, McCulloch CE, Nishida N, Akune T, Yoshimura N, Nakagawa T,
Nevitt MC, Corr M, Lane NE. Variant alleles of the Nakamura K, Tokunaga K, Chung UI, Kawaguchi H.
Wnt antagonist FRZB are determinants of hip shape Transcriptional regulation of endochondral ossification
and modify the relationship between hip shape and by HIF-2alpha during skeletal growth and osteoarthri-
osteoarthritis. Arthritis Rheum 2012;64(5):1457–65. tis development. Nat Med 2010;16(6):678–86.
36. Lodewyckx L, Cailotto F, Thysen S, Luyten FP, Lories 48. Yang S, Kim J, Ryu JH, Oh H, Chun CH, Kim BJ, Min
RJ. Tight regulation of wingless-type signaling in the BH, Chun JS. Hypoxia-inducible factor-2alpha is a
articular cartilage—subchondral bone biomechanical catabolic regulator of osteoarthritic cartilage destruc-
unit: transcriptomics in Frzb-knockout mice. Arthritis tion. Nat Med 2010;16(6):687–93.
Res Ther 2012;14(1):R16. 49. Markway BD, Cho H, Zilberman-Rudenko J, Holden P,
37. Blom AB, Brockbank SM, van Lent PL, van Beuningen McAlinden A, Johnstone B. Hypoxia-inducible factor
HM, Geurts J, Takahashi N, van der Kraan PM, van de 3-alpha expression is associated with the stable chon-
Loo FA, Schreurs BW, Clements K, Newham P, van drocyte phenotype. J Orthopaedic Res 2015;33
den Berg WB. Involvement of the Wnt signaling path- (11):1561–70.
way in experimental and human osteoarthritis: promi- 50. Schipani E, Ryan HE, Didrickson S, Kobayashi T,
nent role of Wnt-induced signaling protein 1. Arthritis Knight M, Johnson RS. Hypoxia in cartilage: HIF-
Rheum 2009;60(2):501–12. 1alpha is essential for chondrocyte growth arrest and
38. Enomoto-Iwamoto M, Nakamura T, Aikawa T, survival. Genes Dev 2001;15(21):2865–76.
Higuchi Y, Yuasa T, Yamaguchi A, Nohno T, Noji S, 51. Choi H, Chun YS, Kim TY, Park JW. HIF-2alpha
Matsuya T, Kurisu K, Koyama E, Pacifici M, Iwamoto enhances beta-catenin/TCF-driven transcription by
M. Hedgehog proteins stimulate chondrogenic cell dif- interacting with beta-catenin. Cancer Res 2010;70
ferentiation and cartilage formation. J Bone Mineral (24):10101–11.
Res 2000;15(9):1659–68. 52. Aref-Eshghi E, Liu M, Harper PE, Dore J, Martin G,
39. Tsumaki N. Bone and calcium update; bone research Furey A, Green R, Rahman P, Zhai G. Overexpression
update. Molecular mechanisms regulating chondrocyte of MMP13 in human osteoarthritic cartilage is asso-
differentiation and hypertrophy. Clin Calcium 2011;21 ciated with the SMAD-independent TGF-beta signal-
(12):113–20. ling pathway. Arthritis Res Ther 2015;17:264.
40. Wei F, Zhou J, Wei X, Zhang J, Fleming BC, Terek R, 53. Adams SB, Setton LA, Bell RD, Easley ME, Huebner JL,
Pei M, Chen Q, Liu T, Wei L. Activation of Indian Stabler T, Kraus VB, Leimer EM, Olson SA, Nettles
hedgehog promotes chondrocyte hypertrophy and DL. Inflammatory cytokines and matrix metalloprotei-
upregulation of MMP-13 in human osteoarthritic car- nases in the synovial fluid after intra-articular ankle
tilage. Osteoarthritis Cartilage 2012;20(7):755–63. fracture. Foot Ankle Int 2015;36(11):1264–71.
41. Nakase T, Miyaji T, Kuriyama K, Tamai N, Horiki M, 54. Vankemmelbeke M, Dekeyser PM, Hollander AP,
Tomita T, Myoui A, Shimada K, Yoshikawa H. Buttle DJ, Demeester J. Characterization of helical
Immunohistochemical detection of parathyroid hor- cleavages in type II collagen generated by matrixins.
mone-related peptide, Indian hedgehog, and patched Biochem J 1998;330(Pt 2):633–40.
in the process of endochondral ossification in the 55. Abula K, Muneta T, Miyatake K, Yamada J, Matsukura
human. Histochem Cell Biol 2001;116(3):277–84. Y, Inoue M, Sekiya I, Graf D, Economides AN, Rosen
42. Pelosi M, Lazzarano S, Thoms BL, Murphy CL. V, et al. Elimination of BMP7 from the developing
Parathyroid hormone-related protein is induced by limb mesenchyme leads to articular cartilage degenera-
hypoxia and promotes expression of the differentiated tion and synovial inflammation with increased age.
phenotype of human articular chondrocytes. Clin Sci FEBS Lett 2015;589(11):1240–48.
2013;125(10):461–70. 56. Little CB, Fosang AJ. Is cartilage matrix breakdown an
43. Koyama E, Ochiai T, Rountree RB, Kingsley DM, appropriate therapeutic target in osteoarthritis—
Enomoto-Iwamoto M, Iwamoto M, Pacifici M. insights from studies of aggrecan and collagen proteo-
Synovial joint formation during mouse limb skeleto- lysis? Curr Drug Targets 2010;11(5):561–75.
genesis: roles of Indian hedgehog signaling. Ann NY 57. Sassi N, Laadhar L, Allouche M, Zandieh-Doulabi B,
Acad Sci 2007;1116:100–12. Hamdoun M, Klein-Nulend J, Makni S, Sellami S. Wnt
44. Zhou J, Chen Q, Lanske B, Fleming BC, Terek R, Wei X, signaling is involved in human articular chondrocyte
Zhang G, Wang S, Li K, Wei L. Disrupting the Indian de-differentiation in vitro. Biotech Histochem 2014;89
hedgehog signaling pathway in vivo attenuates surgically (1):29–40.
induced osteoarthritis progression in Col2a1-CreERT2; 58. Zhou G, Jiang X, Zhang H, Lu Y, Liu A, Ma X, Yang
Ihhfl/fl mice. Arthritis Res Ther 2014;16(1):R11. G, Yang R, Shen H, Zheng J, Hu Y, Yang X, Zhang
45. Saito T, Kawaguchi H. HIF-2alpha as a possible ther- WJ, Xie Z. Zbtb20 regulates the terminal differentia-
apeutic target of osteoarthritis. Osteoarthritis Cartilage tion of hypertrophic chondrocytes via repression of
2010;18(12):1552–56. Sox9. Development 2015;142(2):385–93.
46. Schodel J, Grampp S, Maher ER, Moch H, Ratcliffe PJ, 59. Brew CJ, Clegg PD, Boot-Handford RP, Andrew JG,
Russo P, Mole DR. Hypoxia, hypoxia-inducible Hardingham T. Gene expression in human chondro-
cytes in late osteoarthritis is changed in both fibrillated
RECENT MOLECULAR MECHANISMS OF OSTEOARTHRITIS 257

and intact cartilage without evidence of generalised 72. Scanzello CR, McKeon B, Swaim BH, DiCarlo E,
chondrocyte hypertrophy. Ann Rheumatic Dis Asomugha EU, Kanda V, Nair A, Lee DM, Richmond
2010;69(1):234–40. JC, Katz JN, Crow MK, Goldring SR. Synovial inflamma-
60. Saito T, Ikeda T, Nakamura K, Chung UI, Kawaguchi tion in patients undergoing arthroscopic meniscectomy:
H. S100A1 and S100B, transcriptional targets of SOX molecular characterization and relationship to symptoms.
trio, inhibit terminal differentiation of chondrocytes. Arthritis Rheum 2011;63(2):391–400.
EMBO Rep 2007;8(5):504–9. 73. Sowers M, Karvonen-Gutierrez CA, Jacobson JA, Jiang
61. Wan R, Hu J, Zhou Q, Wang J, Liu P, Wei Y. Y, Yosef M. Associations of anatomical measures from
Application of co-expressed genes to articular cartilage: MRI with radiographically defined knee osteoarthritis
new hope for the treatment of osteoarthritis (review). score, pain, and physical functioning. J Bone Joint Surg
Mol Med Rep 2012;6(1):16–8. Am Volume 2011;93(3):241–51.
62. Nandadasa S, Foulcer S, Apte SS. The multiple, com- 74. Turner DE, Hyslop E, Barn R, McInnes IB, Steultjens
plex roles of versican and its proteolytic turnover by MP, Woodburn J. Metatarsophalangeal joint pain in
ADAMTS proteases during embryogenesis. Matrix Biol psoriatic arthritis: a cross-sectional study. Rheumatol
2014;35:34–41. (Oxford, England) 2014;53(4):737–40.
63. Moncada-Pazos A, Obaya AJ, Viloria CG, Lopez-Otin 75. Benito MJ, Veale DJ, FitzGerald O, van den Berg WB,
C, Cal S. The nutraceutical flavonoid luteolin inhibits Bresnihan B. Synovial tissue inflammation in early and
ADAMTS-4 and ADAMTS-5 aggrecanase activities. J late osteoarthritis. Ann Rheumatic Dis 2005;64
Mol Med (Berlin, Germany) 2011;89(6):611–9. (9):1263–67.
64. Song RH, Tortorella MD, Malfait AM, Alston JT, Yang 76. Ene R, Sinescu RD, Ene P, Cirstoiu MM, Cirstoiu FC.
Z, Arner EC, Griggs DW. Aggrecan degradation in Synovial inflammation in patients with different stages
human articular cartilage explants is mediated by of knee osteoarthritis. Romanian J Morphol Embryol
both ADAMTS-4 and ADAMTS-5. Arthritis Rheum [Revue Roumaine de Morphologie et Embryologie]
2007;56(2):575–85. 2015;56(1):169–73.
65. Demircan K, Topcu V, Takigawa T, Akyol S, Yonezawa 77. Konttinen YT, Pajarinen J, Takakubo Y, Gallo J, Nich
T, Ozturk G, Ugurcu V, Hasgul R, Yigitoglu MR, Akyol C, Takagi M, Goodman SB. Macrophage polarization
O. ADAMTS4 and ADAMTS5 knockout mice are pro- and activation in response to implant debris: influence
tected from versican but not aggrecan or brevican pro- by “particle disease” and “ion disease”. J Long-term Eff
teolysis during spinal cord injury. Biomed Res Int Med Implants 2014;24(4):267–81.
2014;2014:693746. 78. Jiang X, Sato T, Yao Z, Keeney M, Pajarinen J, Lin TH,
66. Fotinos-Hoyer AK, Guermazi A, Jara H, Eckstein F, Loi F, Egashira K, Goodman S, Yang F. Local delivery
Ozonoff A, Khard H, Norbash A, Bohndorf K, Roemer of mutant CCL2 protein-reduced orthopaedic implant
FW. Assessment of synovitis in the osteoarthritic knee: wear particle-induced osteolysis and inflammation in
comparison between manual segmentation, semiauto- vivo. J Orthopaedic Res: Off Publ Orthopaedic Res Soc
mated segmentation, and semiquantitative assessment 2016;34(1):58–64.
using contrast-enhanced fat-suppressed T1-weighted 79. Blom AB, van Lent PL, Libregts S, Holthuysen AE, van
MRI. Magn Reson Med 2010;64(2):604–9. der Kraan PM, van Rooijen N, van den Berg WB.
67. Krenn V, Morawietz L, Burmester GR, Kinne RW, Crucial role of macrophages in matrix metalloprotei-
Mueller-Ladner U, Muller B, Haupl T. Synovitis score: nase-mediated cartilage destruction during experimental
discrimination between chronic low-grade and high- osteoarthritis: involvement of matrix metalloproteinase
grade synovitis. Histopathology 2006;49(4):358–64. 3. Arthritis Rheum 2007;56(1):147–57.
68. Slansky E, Li J, Haupl T, Morawietz L, Krenn V, Pessler 80. van Lent PL, Blom AB, van der Kraan P, Holthuysen
F. Quantitative determination of the diagnostic accu- AE, Vitters E, van Rooijen N, Smeets RL, Nabbe KC,
racy of the synovitis score and its components. van den Berg WB. Crucial role of synovial lining
Histopathology 2010;57(3):436–43. macrophages in the promotion of transforming growth
69. Pessler F, Dai L, Diaz-Torne C, Gomez-Vaquero C, factor beta-mediated osteophyte formation. Arthritis
Paessler ME, Zheng DH, Einhorn E, Range U, Rheum 2004;50(1):103–11.
Scanzello C, Schumacher HR. The synovitis of “non- 81. Bondeson J, Wainwright SD, Lauder S, Amos N,
inflammatory” orthopaedic arthropathies: a quantita- Hughes CE. The role of synovial macrophages and
tive histological and immunohistochemical analysis. macrophage-produced cytokines in driving aggreca-
Ann Rheumatic Dis 2008;67(8):1184–7. nases, matrix metalloproteinases, and other destructive
70. Wenham CY, Balamoody S, Grainger AJ, Hensor EM, and inflammatory responses in osteoarthritis. Arthritis
Draycott S, Hodgson R, Conaghan PG. The respon- Res Ther 2006;8(6):R187.
siveness of novel, dynamic, contrast-enhanced mag- 82. Takano S, Uchida K, Miyagi M, Inoue G, Aikawa J,
netic resonance measures of total knee synovitis after Fujimaki H, Minatani A, Sato M, Iwabuchi K, Takaso
intra-articular corticosteroid for painful osteoarthritis. M. Synovial macrophage-derived IL1B regulates the
Osteoarthritis Cartilage 2014;22(10):1614–18. calcitonin receptor in osteoarthritic mice. Clin Exp
71. Baker K, Grainger A, Niu J, Clancy M, Guermazi A, Crema Immunol 2016;183(1):143–9.
M, Hughes L, Buckwalter J, Wooley A, Nevitt M, Felson 83. Han SA, Lee S, Seong SC, Lee MC. Effects of CD14
DT. Relation of synovitis to knee pain using contrast- macrophages and proinflammatory cytokines on chon-
enhanced MRIs. Annals Rheumatic Dis 2010;69 drogenesis in osteoarthritic synovium-derived stem
(10):1779–83. cells. Tissue Eng Part A 2014;20(19–20):2680–91.
258 Y. WEI AND L. BAI

84. Fahy N, de Vries-van Melle ML, Lehmann J, Wei W, 96. Tarkowski A, Trollmo C, Seifert PS, Hansson GK.
Grotenhuis N, Farrell E, van der Kraan PM, Murphy Expression of decay-accelerating factor on synovial
JM, Bastiaansen-Jenniskens YM, van Osch GJ. Human lining cells in inflammatory and degenerative arthri-
osteoarthritic synovium impacts chondrogenic differ- tides. Rheumatology Int 1992;12(5):201–5.
entiation of mesenchymal stem cells via macrophage 97. Geyer M, Grassel S, Straub RH, Schett G, Dinser R,
polarisation state. Osteoarthritis Cartilage 2014;22 Grifka J, Gay S, Neumann E, Muller-Ladner U.
(8):1167–75. Differential transcriptome analysis of intraarticular
85. Uchida K, Satoh M, Inoue G, Onuma K, Miyagi M, lesional vs intact cartilage reveals new candidate genes
Iwabuchi K, Takaso M. CD11c(+) macrophages and in osteoarthritis pathophysiology. Osteoarthritis
levels of TNF-alpha and MMP-3 are increased in syno- Cartilage 2009;17(3):328–35.
vial and adipose tissues of osteoarthritic mice with 98. Gobezie R, Kho A, Krastins B, Sarracino DA, Thornhill
hyperlipidaemia. Clin Exp Immunol 2015;180(3):551–9. TS, Chase M, Millett PJ, Lee DM. High abundance
86. Moradi B, Rosshirt N, Tripel E, Kirsch J, Barie A, synovial fluid proteome: distinct profiles in health
Zeifang F, Gotterbarm T, Hagmann S. and osteoarthritis. Arthritis Res Ther 2007;9(2):R36.
Unicompartmental and bicompartmental knee osteoar- 99. Mateos J, Lourido L, Fernandez-Puente P, Calamia V,
thritis show different patterns of mononuclear cell Fernandez-Lopez C, Oreiro N, Ruiz-Romero C,
infiltration and cytokine release in the affected joints. Blanco FJ. Differential protein profiling of synovial
Clinical Exp Immunol 2015;180(1):143–54. fluid from rheumatoid arthritis and osteoarthritis
87. Kumar A, Shamsuddin N. Retinal Muller glia initiate patients using LC-MALDI TOF/TOF. J Proteomics
innate response to infectious stimuli via toll-like recep- 2012;75(10):2869–78.
tor signaling. PloS One 2012;7(1):e29830. 100. Fernandez-Puente P, Mateos J, Fernandez-Costa C,
88. Scanzello CR, Plaas A, Crow MK. Innate immune Oreiro N, Fernandez-Lopez C, Ruiz-Romero C,
system activation in osteoarthritis: is osteoarthritis a Blanco FJ. Identification of a panel of novel serum
chronic wound? Curr Opin Rheumatol 2008;20 osteoarthritis biomarkers. J Proteome Res 2011;10
(5):565–72. (11):5095–101.
89. Ishijima M, Watari T, Naito K, Kaneko H, Futami I, 101. Rosenthal AK, Gohr CM, Ninomiya J, Wakim BT.
Yoshimura-Ishida K, Tomonaga A, Yamaguchi H, Proteomic analysis of articular cartilage vesicles from
Yamamoto T, Nagaoka I, Kurosawa H, Poole RA, normal and osteoarthritic cartilage. Arthritis Rheum
Kaneko K. Relationships between biomarkers of car- 2011;63(2):401–11.
tilage, bone, synovial metabolism and knee pain pro- 102. Cantatore FP, Benazzo F, Ribatti D, Lapadula G,
vide insights into the origins of pain in early knee D’Amico S, Tursi A, Pipitone V. Early alteration of
osteoarthritis. Arthritis Res Ther 2011;13(1):R22. synovial membrane in osteoarthrosis. Clin Rheumatol
90. Tamaki Y, Takakubo Y, Hirayama T, Konttinen YT, 1988;7(2):214–9.
Goodman SB, Yamakawa M, Takagi M: Expression of 103. Breitner S, Storkel S, Reichel W, Loos M. Complement
Toll-like receptors and their signaling pathways in components C1q, C1r/C1s, and C1INH in rheumatoid
rheumatoid synovitis. J Rheumatol 2011;38(5):810–20. arthritis. Correlation of in situ hybridization and
91. Carrion M, Juarranz Y, Perez-Garcia S, Jimeno R, northern blot results with function and protein con-
Pablos JL, Gomariz RP, Gutierrez-Canas I. RNA centration in synovium and primary cell cultures.
sensors in human osteoarthritis and rheumatoid Arthritis Rheum 1995;38(4):492–8.
arthritis synovial fibroblasts: immune regulation by 104. Sjoberg A, Onnerfjord P, Morgelin M, Heinegard D,
vasoactive intestinal peptide. Arthritis Rheum Blom AM. The extracellular matrix and inflammation:
2011;63(6):1626–36. fibromodulin activates the classical pathway of comple-
92. Gay NJ, Symmons MF, Gangloff M, Bryant CE. ment by directly binding C1q. J Biol Chem 2005;280
Assembly and localization of Toll-like receptor signal- (37):32301–08.
ling complexes. Nat Rev Immunol 2014;14(8):546–58. 105. Happonen KE, Saxne T, Aspberg A, Morgelin M,
93. Midwood K, Sacre S, Piccinini AM, Inglis J, Trebaul A, Heinegard D, Blom AM. Regulation of complement
Chan E, Drexler S, Sofat N, Kashiwagi M, Orend G, by cartilage oligomeric matrix protein allows for a
Brennan F, Foxwell B. Tenascin-C is an endogenous novel molecular diagnostic principle in rheumatoid
activator of Toll-like receptor 4 that is essential for arthritis. Arthritis Rheum 2010;62(12):3574–83.
maintaining inflammation in arthritic joint disease. 106. Sjoberg AP, Manderson GA, Morgelin M, Day AJ,
Nat Med 2009;15(7):774–80. Heinegard D, Blom AM. Short leucine-rich glycopro-
94. Wang X, Hunter D, Xu J, Ding C. Metabolic triggered teins of the extracellular matrix display diverse patterns
inflammation in osteoarthritis. Osteoarthritis Cartilage of complement interaction and activation. Mol
2015;23(1):22–30. Immunol 2009;46(5):830–9.
95. Ritter SY, Subbaiah R, Bebek G, Crish J, Scanzello CR, 107. Kalchishkova N, Furst CM, Heinegard D, Blom AM.
Krastins B, Sarracino D, Lopez MF, Crow MK, Aigner NC4 Domain of cartilage-specific collagen IX inhibits
T, Goldring MB, Goldring SR, Lee DM, Gobezie R, complement directly due to attenuation of membrane
Aliprantis AO. Proteomic analysis of synovial fluid attack formation and indirectly through binding and
from the osteoarthritic knee: comparison with tran- enhancing activity of complement inhibitors C4B-
scriptome analyses of joint tissues. Arthritis Rheum binding protein and factor H. J Biol Chem 2011;286
2013;65(4):981–92. (32):27915–26.
RECENT MOLECULAR MECHANISMS OF OSTEOARTHRITIS 259

108. Lepus CM, Song JJ, Wang Q, Wagner CA, Lindstrom progression of osteoarthritis by inducing macrophage
TM, Chu CR, Sokolove J, Leung LL, Robinson WH. inflammatory protein-1gamma. Osteoarthritis
Brief report: carboxypeptidase B serves as a protective Cartilage 2011;19(6):728–36.
mediator in osteoarthritis. Arthritis Rheumatology 122. Hayden MS, Ghosh S. Signaling to NF-kappaB. Genes
2014;66(1):101–6. Dev 2004;18(18):2195–224.
109. Sellam J, Berenbaum F. The role of synovitis in patho- 123. Marcu KB, Otero M, Olivotto E, Borzi RM, Goldring
physiology and clinical symptoms of osteoarthritis. Nat MB. NF-kappaB signaling: multiple angles to target
Rev Rheumatology 2010;6(11):625–35. OA. Curr Drug Targets 2010;11(5):599–613.
110. Kapoor M, Martel-Pelletier J, Lajeunesse D, Pelletier 124. Steenvoorden MM, Huizinga TW, Verzijl N, Bank RA,
JP, Fahmi H. Role of proinflammatory cytokines in the Ronday HK, Luning HA, Lafeber FP, Toes RE,
pathophysiology of osteoarthritis. Nat Rev DeGroot J. Activation of receptor for advanced glyca-
Rheumatology 2011;7(1):33–42. tion end products in osteoarthritis leads to increased
111. Litherland GJ, Hui W, Elias MS, Wilkinson DJ, Watson stimulation of chondrocytes and synoviocytes. Arthritis
S, Huesa C, Young DA, Rowan AD. Glycogen synthase Rheum 2006;54(1):253–63.
kinase 3 inhibition stimulates human cartilage destruc- 125. Wang SN, Xie GP, Qin CH, Chen YR, Zhang KR, Li X,
tion and exacerbates murine osteoarthritis. Arthritis Wu Q, Dong WQ, Yang J, Yu B. Aucubin prevents
Rheumatology 2014;66(8):2175–87. interleukin-1 beta induced inflammation and cartilage
112. Chevalier X, Giraudeau B, Conrozier T, Marliere J, matrix degradation via inhibition of NF-kappaB signal-
Kiefer P, Goupille P. Safety study of intraarticular ing pathway in rat articular chondrocytes. Int
injection of interleukin 1 receptor antagonist in Immunopharmacol 2015;24(2):408–15.
patients with painful knee osteoarthritis: a multicenter 126. Englund M, Guermazi A, Lohmander LS. The menis-
study. J Rheumatology 2005;32(7):1317–23. cus in knee osteoarthritis. Rheumatic Dis Clin North
113. Pazar B, Ea HK, Narayan S, Kolly L, Bagnoud N, America 2009;35(3):579–90.
Chobaz V, Roger T, Liote F, So A, Busso N. Basic 127. Englund M, Guermazi A, Roemer FW, Aliabadi P,
calcium phosphate crystals induce monocyte/macro- Yang M, Lewis CE, Torner J, Nevitt MC, Sack B,
phage IL-1beta secretion through the NLRP3 inflam- Felson DT. Meniscal tear in knees without surgery
masome in vitro. J Immunol (Baltimore, Md: 1950) and the development of radiographic osteoarthritis
2011;186(4):2495–502. among middle-aged and elderly persons: the
114. Jin C, Frayssinet P, Pelker R, Cwirka D, Hu B, Vignery Multicenter Osteoarthritis Study. Arthritis Rheum
A, Eisenbarth SC, Flavell RA. NLRP3 inflammasome 2009;60(3):831–39.
plays a critical role in the pathogenesis of hydroxyapa- 128. Roemer FW, Kwoh CK, Hannon MJ, Hunter DJ,
tite-associated arthropathy. Proc National Acad Sci US Eckstein F, Fujii T, Boudreau RM, Guermazi A. What
A 2011;108(36):14867–72. comes first? Multitissue involvement leading to radio-
115. Schlaak JF, Pfers I, Meyer Zum Buschenfelde KH, graphic osteoarthritis: magnetic resonance imaging-
Marker-Hermann E. Different cytokine profiles in the based trajectory analysis over four years in the osteoar-
synovial fluid of patients with osteoarthritis, rheuma- thritis initiative. Arthritis Rheumatology 2015;67
toid arthritis and seronegative spondylarthropathies. (8):2085–96.
Clin Exp Rheumatology 1996;14(2):155–62. 129. Lawrence JT, Birmingham J, Toth AP. Emerging
116. Zhang X, Zhu Y, Chen X, Zhang Y, Zhang Y, Jia Y, ideas: prevention of posttraumatic arthritis through
Wang H, Liu Y, Xiao L. Baicalein ameliorates inflam- interleukin-1 and tumor necrosis factor-alpha inhibi-
matory-related apoptotic and catabolic phenotypes in tion. Clin Orthopaedics Relat Res 2011;469
human chondrocytes. Int Immunopharmacol 2014;21 (12):3522–26.
(2):301–8. 130. Scanzello CR, Umoh E, Pessler F, Diaz-Torne C, Miles
117. Elson CJ, Mortuza FY, Perry MJ, Warnock MG, Webb T, Dicarlo E, Potter HG, Mandl L, Marx R, Rodeo S,
GR, Westacott CI. Cytokines and focal loss of cartilage Goldring SR, Crow MK. Local cytokine profiles in knee
in osteoarthritis. Br J Rheumatology 1998;37(1):106–7. osteoarthritis: elevated synovial fluid interleukin-15
118. Wojdasiewicz P, Poniatowski LA, Szukiewicz D. The differentiates early from end-stage disease.
role of inflammatory and anti-inflammatory cytokines Osteoarthritis Cartilage 2009;17(8):1040–48.
in the pathogenesis of osteoarthritis. Mediat Inflamm 131. Hellio Le Graverand MP, Vignon E, Otterness IG, Hart
2014;2014:561459. DA. Early changes in lapine menisci during osteoar-
119. Magnano MD, Chakravarty EF, Broudy C, Chung L, thritis development. Part II: molecular alterations.
Kelman A, Hillygus J, Genovese MC. A pilot study of Osteoarthritis Cartilage 2001;9(1):65–72.
tumor necrosis factor inhibition in erosive/inflamma- 132. McNulty AL, Miller MR, O’Connor SK, Guilak F. The
tory osteoarthritis of the hands. J Rheumatology effects of adipokines on cartilage and meniscus catabo-
2007;34(6):1323–27. lism. Connective Tissue Res 2011;52(6):523–33.
120. Amin AR, Islam AB. Genomic analysis and differential 133. Fuller ES, Smith MM, Little CB, Melrose J. Zonal
expression of HMG and S100A family in human differences in meniscus matrix turnover and cytokine
arthritis: upregulated expression of chemokines, IL-8 response. Osteoarthritis Cartilage 2012;20(1):49–59.
and nitric oxide by HMGB1. DNA Cell Biol 2014;33 134. Mitroulis I, Alexaki VI, Kourtzelis I, Ziogas A,
(8):550–65. Hajishengallis G, Chavakis T. Leukocyte integrins: role
121. Shen PC, Wu CL, Jou IM, Lee CH, Juan HY, Lee PJ, in leukocyte recruitment and as therapeutic targets in
Chen SH, Hsieh JL. T helper cells promote disease inflammatory disease. Pharmacol Ther 2015;147:123–35.
260 Y. WEI AND L. BAI

135. Schett G, Kiechl S, Bonora E, Zwerina J, Mayr A, 151. Racine J, Aaron RK. Pathogenesis and epidemiology of
Axmann R, Weger S, Oberhollenzer F, Lorenzini R, osteoarthritis. Rhode Island Med J (2013) 2013;96
Willeit J. Vascular cell adhesion molecule 1 as a pre- (3):19–22.
dictor of severe osteoarthritis of the hip and knee 152. Gabusi E, Manferdini C, Grassi F, Piacentini A, Cattini L,
joints. Arthritis Rheum 2009;60(8):2381–89. Filardo G, Lambertini E, Piva R, Zini N, Facchini A,
136. Zhuo Q, Yang W, Chen J, Wang Y. Metabolic syn- Lisignoli G. Extracellular calcium chronically induced
drome meets osteoarthritis. Nat Rev Rheumatology human osteoblasts effects: specific modulation of osteo-
2012;8(12):729–37. calcin and collagen type XV. J Cell Physiol 2012;227
137. Gregor MF, Hotamisligil GS. Inflammatory mechan- (8):3151–61.
isms in obesity. Annu Rev Immunol 2011;29:415–45. 153. Bailey AJ, Sims TJ, Knott L. Phenotypic expression of
138. Andersen CJ, Murphy KE, Fernandez ML. Impact of osteoblast collagen in osteoarthritic bone: production
obesity and metabolic syndrome on immunity. Adv of type I homotrimer. Int J Biochem Cell Biol 2002;34
Nutr (Bethesda, Md) 2016;7(1):66–75. (2):176–82.
139. Ferraz-Amaro I, Gonzalez-Juanatey C, Lopez-Mejias R, 154. Sakao K, Takahashi KA, Arai Y, Saito M, Honjo K,
Riancho-Zarrabeitia L, Gonzalez-Gay MA. Metabolic Hiraoka N, Asada H, Shin-Ya M, Imanishi J, Mazda O,
syndrome in rheumatoid arthritis. Mediat Inflamm Kubo T. Osteoblasts derived from osteophytes produce
2013;2013:710928. interleukin-6, interleukin-8, and matrix metalloprotei-
140. Hotamisligil GS. Inflammation and metabolic disor- nase-13 in osteoarthritis. J Bone Mineral Metab
ders. Nature 2006;444(7121):860–7. 2009;27(4):412–23.
141. Monteiro R, Azevedo I. Chronic inflammation in obe- 155. Sanchez C, Deberg MA, Bellahcene A, Castronovo V,
sity and the metabolic syndrome. Mediat Inflamm Msika P, Delcour JP, Crielaard JM, Henrotin YE.
2010;2010:289645. Phenotypic characterization of osteoblasts from the
142. Emanuela F, Grazia M, Marco de R, Maria Paola L, sclerotic zones of osteoarthritic subchondral bone.
Giorgio F, Marco B. Inflammation as a Link between Arthritis Rheum 2008;58(2):442–55.
Obesity and Metabolic Syndrome. J Nutr Metab 156. Sanchez C, Pesesse L, Gabay O, Delcour JP, Msika P,
2012;2012:476380. Baudouin C, Henrotin YE. Regulation of subchondral
143. Zeyda M, Farmer D, Todoric J, Aszmann O, Speiser M, bone osteoblast metabolism by cyclic compression.
Gyori G, Zlabinger GJ, Stulnig TM. Human adipose tissue Arthritis Rheum 2012;64(4):1193–203.
macrophages are of an anti-inflammatory phenotype but 157. Sanchez C, Horcajada MN, Membrez Scalfo F, Ameye
capable of excessive pro-inflammatory mediator produc- L, Offord E, Henrotin Y. Carnosol inhibits pro-inflam-
tion. Int J Obesity (2005) 2007;31(9):1420–28. matory and catabolic mediators of cartilage breakdown
144. Clockaerts S, Bastiaansen-Jenniskens YM, Runhaar J, in human osteoarthritic chondrocytes and mediates
Van Osch GJ, Van Offel JF, Verhaar JA, De Clerck LS, cross-talk between subchondral bone osteoblasts and
Somville J. The infrapatellar fat pad should be consid- chondrocytes. PloS One 2015;10(8):e0136118.
ered as an active osteoarthritic joint tissue: a narrative 158. Kushlinskii NE, Timofeev YS, Solov’ev YN, Gerstein
review. Osteoarthritis Cartilage 2010;18(7):876–82. ES, Lyubimova NV, Bulycheva IV. Components of
145. Han W, Cai S, Liu Z, Jin X, Wang X, Antony B, Cao Y, the RANK/RANKL/OPG system, IL-6, IL-8, IL-16,
Aitken D, Cicuttini F, Jones G, Ding C. Infrapatellar fat MMP-2, and calcitonin in the sera of patients
pad in the knee: is local fat good or bad for knee with bone tumors. Bull Exp Biol Med 2014;157
osteoarthritis? Arthritis Research Ther 2014;16(4):R145. (4):520–3.
146. Pesesse L, Sanchez C, Henrotin Y. Osteochondral plate 159. Di Francesco S, Castellan P, Manco R, Tenaglia RL.
angiogenesis: a new treatment target in osteoarthritis. Reciprocal cross-talk between Prostaglandin E2 and
Joint, Bone, Spine: revue du rhumatisme 2011;78 bone in prostate cancer: a current review. Cent Eur J
(2):144–9. Urology 2011;64(4):201–4.
147. Anderson-MacKenzie JM, Quasnichka HL, Starr RL, 160. Kaspiris A, Khaldi L, Grivas TB, Vasiliadis E, Kouvaras
Lewis EJ, Billingham ME, Bailey AJ. Fundamental sub- I, Dagkas S, Chronopoulos E, Papadimitriou E.
chondral bone changes in spontaneous knee osteoar- Subchondral cyst development and MMP-1 expression
thritis. Int J Biochem Cell Biol 2005;37(1):224–36. during progression of osteoarthritis: an immunohisto-
148. Klose-Jensen R, Hartlev LB, Boel LW, Laursen MB, chemical study. Orthopaedics Traumatology, Surg Res:
Stengaard-Pedersen K, Keller KK, Hauge EM. OTSR 2013;99(5):523–29.
Subchondral bone turnover, but not bone volume, is 161. Mapp PI, Walsh DA. Mechanisms and targets of angio-
increased in early stage osteoarthritic lesions in the genesis and nerve growth in osteoarthritis. Nat Rev
human hip joint. Osteoarthritis Cartilage 2015;23 Rheumatology 2012;8(7):390–8.
(12):2167–73. 162. Binks DA, Gravallese EM, Bergin D, Hodgson RJ, Tan
149. Thambyah A, Broom N. On new bone formation in the AL, Matzelle MM, McGonagle D, Radjenovic A. Role
pre-osteoarthritic joint. Osteoarthritis Cartilage of vascular channels as a novel mechanism for sub-
2009;17(4):456–63. chondral bone damage at cruciate ligament entheses in
150. Milgram JW. Morphologic alterations of the subchon- osteoarthritis and inflammatory arthritis. Ann
dral bone in advanced degenerative arthritis. Clin Rheumatic Dis 2015;74(1):196–203.
Orthopaedics Relat Res 1983;173:293–312. 163. Masuko K, Murata M, Beppu M, Nakamura H, Kato T,
Yudoh K: Sphingosine-1-phosphate modulates expres-
sion of vascular endothelial growth factor in human
RECENT MOLECULAR MECHANISMS OF OSTEOARTHRITIS 261

articular chondrocytes: a possible new role in arthritis. 168. Bouaziz W, Funck-Brentano T, Lin H, Marty C, Ea
Int J Rheumatic Dis 2012;15(4):366–73. HK, Hay E, Cohen-Solal M. Loss of sclerostin pro-
164. Lories RJ, Corr M, Lane NE. To Wnt or not to Wnt: motes osteoarthritis in mice via beta-catenin-depen-
the bone and joint health dilemma. Nat Rev dent and -independent Wnt pathways. Arthritis Res
Rheumatology 2013;9(6):328–39. Ther 2015;17:24.
165. Baron R, Kneissel M. WNT signaling in bone home- 169. Bouvard B, Abed E, Yelehe-Okouma M, Bianchi A,
ostasis and disease: from human mutations to treat- Mainard D, Netter P, Jouzeau JY, Lajeunesse D,
ments. Nat Med 2013;19(2):179–92. Reboul P. Hypoxia and vitamin D differently contribute
166. Golovchenko S, Hattori T, Hartmann C, Gebhardt M, to leptin and dickkopf-related protein 2 production in
Gebhard S, Hess A, Pausch F, Schlund B, von der Mark human osteoarthritic subchondral bone osteoblasts.
K. Deletion of beta catenin in hypertrophic growth Arthritis Res Ther 2014;16(5):459.
plate chondrocytes impairs trabecular bone formation. 170. Zhen G, Wen C, Jia X, Li Y, Crane JL, Mears SC, Askin
Bone 2013;55(1):102–12. FB, Frassica FJ, Chang W, Yao J, Carrino JA, Cosgarea
167. Funck-Brentano T, Bouaziz W, Marty C, Geoffroy A, Artemov D, Chen Q, Zhao Z, Zhou X, Riley L,
V, Hay E, Cohen-Solal M. Dkk-1-mediated inhibi- Sponseller P, Wan M, Lu WW, Cao X. Inhibition of
tion of Wnt signaling in bone ameliorates osteoar- TGF-beta signaling in mesenchymal stem cells of sub-
thritis in mice. Arthritis Rheumatology 2014;66 chondral bone attenuates osteoarthritis. Nat Med
(11):3028–39. 2013;19(6):704–12.

You might also like