Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 Oct 1.
Published in final edited form as: Osteoarthritis Cartilage. 2013 Jun 14;21(10):1436–1442. doi: 10.1016/j.joca.2013.05.020

Osteoarthritis Year in Review 2013: Biology

Richard F Loeser 1
PMCID: PMC3779513  NIHMSID: NIHMS504076  PMID: 23774472

Abstract

The purpose of this review was to present highlights from the published literature on the topic of the biology of osteoarthritis (OA). A PubMed search was conducted in order to locate original research manuscripts published since the last OARSI meeting in 2012. From review of the published literature, common themes emerged as active areas of research over the past year including studies in the areas of epigenetics, Wnt signaling, the role of inflammatory pathways in OA, lubricin, fibroblast growth factor signaling, and studies on OA biology in bone. Key findings in these areas were summarized and implications for future therapies were discussed.

Keywords: inflammation, signaling, growth factors, bone, cartilage, meniscus, synovium

Introduction

Research into the basic biology of osteoarthritis (OA) continues to grow with many investigators focused on elucidating mechanisms relevant to cartilage matrix destruction. However, an accumulating number of studies have reported findings in other joint tissues affected by OA including bone, synovium, ligaments, and the menisci. Since OA is a disease of the joint as an organ, knowledge of how these tissues interact to result in OA will be important and could lead to new approaches for the treatment. This review attempts to capture and summarize key findings in the biology of OA published since the last OARSI meeting in April 2012 up to the end of March 2013.

A Pubmed search of English language articles from April 1, 2012 to March 24, 2013 under the search term of osteoarthritis with MESH terms of embryology, enzymology, etiology, immunology, metabolism, pathology, physiology, and physiopathology returned 843 published articles. The titles and selected abstracts from this list were reviewed and publications relevant to the biology of OA were collected. In addition, selected journals in the field were cross-checked for additional articles of interest that were not picked up in the Pubmed search. Articles more relevant to cartilage tissue engineering were not included in the review in order to focus on the biology of OA.

Many of the topic areas covered by Peter van der Kraan in the 2012 review1, such as epigenetics (in particular micro-RNA), Wnt signaling, autophagy, inflammation, obesity/metabolic syndrome, aging, and the concept of OA as a disease of the joint as an organ, continued to be active areas of investigation over the past year. From these topic areas, epigenetics and Wnt signaling were particularly active areas and so these will be discussed. In addition, the role of inflammatory pathways in OA, including those involved in innate immune responses, was chosen to highlight along with selected publications on lubricin, fibroblast growth factor (FGF) signaling, and studies on OA biology in bone. It is not possible to cover all of the excellent work being done in the field and so apologies to those whose work was not included.

Epigenetics

Epigenetic regulation of gene transcription allows cells to alter and fine tune gene expression in response to external cues. Unlike genetic modifications which require very long periods of evolutionary pressure to occur, organisms can use epigenetic modifications to more rapidly respond to changes in the environment. Individual cells and tissues can likewise use epigenetic modifications to alter gene expression to respond to changes in their microenvironment. Many epigenetic modifications can be maintained in cultured cells. It has been observed for quite some time that chondrocytes removed from an osteoarthritic joint and placed in culture will maintain an OA-like phenotype for a week or more, expressing genes seen in OA despite the fact that the cells are no longer in the presence of the altered milieu of the OA joint. This phenomenon could be explained by epigenetic changes in the cell, including DNA methylation, histone methylation and acetylation, and micro (mi) RNAs, which regulate expression of the genes related to the OA-phenotype.

MMP-13 is a critical enzyme that mediates cartilage matrix destruction in OA and which appears to be under epigenetic regulation. Increased MMP-13 expression by human OA chondrocytes was associated with demethylation of CpG sites in the MMP-13 promoter2. A site at -104 bp was studied further and found to be a site for binding of the cAMP response element-binding (CREB) when the site was demethylated. In another MMP-13 promoter study, demethylation at -110 bp and -299bp in human chondrocytes was associated with increased levels of MMP-13 gene expression3. The same study found similar sites in the IL-1β promoter. Interestingly, binding of HIF-2α, which was recently implicated in promoting OA in mice, to the MMP-13 but not IL-1β promoter, was greater when the two sites were demethylated. CREB was not tested but RUNX2, AP-1, and ELF3 regulation of MMP-13 expression was not altered by methylation status.

Another gene associated with catabolic activity in cartilage is the inducible nitric oxide synthase (iNOS). A site in the NF-κB enhancer region was found to be demethylated in human OA chondrocytes relative to controls and methylation of this region was associated with decreased iNOS expression4. On the anabolic side, methylation of the SOX-9 promoter was found to be increased rather than decreased in human OA cartilage relative to normal controls5. The demethylating agent 5-azacytidine was able to increase SOX-9 expression demonstrating that methylation was inhibitory and this appeared to be mediated by altered binding of the CCAAT-binding factor/nuclear factor-Y and a cAMP response element-binding factor. Histone methylation and acetylation may also regulate SOX-9 promoter activity and an increase in H3K9 and H3K27 trimethylation and a decrease in acetylation at H3K9, 15, 18, 23, and 27 at the SOX-9 promoter was found.

Histone modifications can also regulate catabolic mediators in cartilage. Human chondrocytes induced to express RUNX-2, MMP-3 and ADAMTS-5 by mechanical stress were treated with histone deacetylase (HDAC) inhibitors MS-275 or trichostatin A (TSA)6. The HDAC inhibitors reduced expression of all three genes. Likewise, IL-1-induced expression of MMP-1, MMP-3, and MMP-13 was reduced by treatment of human articular chondrocytes with TSA, MS-275, or valproic acid7. In further support for an in vivo role for histone acetylation in OA, TSA treatment of mice reduced the severity of cartilage damage in the DMM model of OA7. Thus, studies to date strongly implicate a number of catabolic and anabolic factors in OA chondrocytes which are regulated by both methylation and histone modifications.

In addition to studies in cartilage, differences in DNA methylation in OA versus osteoporotic bone, taken from the central part of the femoral head, have been examined8. Out of 241 sites in 228 genes that were differentially methylated, the vast majority (217) were found to have reduced methylation in osteoporotic bone. When these genes were annotated for function, the homeobox superfamily of transcription factors was found to be significantly different between OA and osteoporotic bone. This is of interest given the important role of this family of transcription factors in bone and joint development.

A very active area of epigenetics research in the past year has been the study of miRNAs although which ones might be important in the biology of OA is not completely clear. A microarray analysis of miRNAs present in cultured human normal and OA chondrocytes found 7 miRNAs differentially expressed9. Only one, miR483-5p was increased in OA cells while the others (miR-149, miR-582-3p, miR-1227, miR-634, miR576-5p, and miR-641) were at higher levels in normal chondrocytes. Functional studies were not performed and so it was not clear what role if any these have in OA. Another study using human cartilage noted an age-related increase in miR-199a-3p and miR193b10. In contrast, IL-1β treated human chondrocytes had decreased levels of miR-199a and this was associated with increased expression of cyclooxygenase-2 and subsequent PGE-2 production11. IL-1β treatment of the immortalized C28/I2 chondrocyte line increased expression of miRNA-140 which was found to inhibit MMP-13 expression12. A previous study had shown that deletion of miRNA-140 in mice resulted in a premature OA phenotype in association with increased Adamts5 expression13.

Wnt Signaling and OA

Wnt signaling plays a key role in the development of the joint and recent work supports a role in OA as well. Because Wnt pathways are involved in both cartilage and bone formation that include a role in regulation of chondrocyte hypertrophy in the growth plate, disregulation of Wnt pathways in adult tissues could contribute to the chondrocyte hypertrophy seen in OA and pathologic changes in cartilage and bone. Sclerostin serves as an inhibitor of Wnt signaling in bone and loss of function mutations in sclerostin or inhibition through the use of neutralizing antibodies result in increased bone mass. In addition to bone, Roudier et al14 confirmed earlier reports demonstrating sclerostin was also expressed in articular cartilage, albeit at similar levels in normal and OA human cartilage. However, neither genetic deletion of sclerostin in mice or antibody inhibition in rats altered the development of naturally occurring OA in mice or surgically-induced OA in rats. Interestingly, sclerostin knock-out mice at 12 and 16 months of age did not have more severe cartilage lesions compared to age-matched controls despite significantly increased subchondral bone mass. This finding either argues against a direct effect of increased subchondral bone mass on the overlying cartilage or suggests loss of sclerostin in cartilage might have counter-acted the effect of increased bone.

Dkks 1-4 are also Wnt antagonists and Dkk-1 and -2 were reported to be present in articular cartilage with Dkk-1 also found in human OA synovium15, 16. Dkk-1 was increased while Dkk-2 was decreased in OA cartilage when compared to normal 15. In another study, Dkk-1 and two other Wnt antagonists, Gremelin 1 and Frizzled-related protein, were found to be expressed at higher levels in articular cartilage when compared to growth plate17. These Wnt antagonists were shown to inhibit chondrocyte hypertrophy in vitro suggesting the higher levels in articular cartilage might serve to inhibit hypertrophy. Dkk-1 overexpression in the mouse joint by adenoviral injection or by transgenic overexpression resulted in reduced OA severity in the destabilized medial meniscus (DMM) model 15. However, the opposite effect of Dkk-1 on OA was seen in the rat anterior cruciate ligament transection (ACLT) model where intra-peritoneal injection of Dkk-1 antisense oligonucleotides sufficient to reduce the number of Dkk-1 positive cells in the synovium reduced the severity of OA rather than increasing it16. Dkk-1 was shown to inhibit MMP-13 and ADAMTS-4 expression in chondrocytes in response to the Wnt pathway activator Wnt-3a15 while it promoted production of pro-angiogenic factors and proteinases (ADAMTS-5 and MMP-3) produced by synovial fibroblasts16. If these findings of opposite effects in cartilage and synovium are confirmed, it would suggest that Dkk-1 would not serve as a very good target for treatment of OA when both tissues are affected.

Adding to the complexity of the role of Wnt signaling in OA, a study of the Wnt-1-inducible signaling protein 3/CCN6 found that it was highly expressed in human OA cartilage compared to normal cartilage and in vitro experiments found that it down-regulated ADAMTS-5 expression while up-regulating MMP-1018. Activation of Wnt/β-catenin signaling in primary human chondrocytes inhibited basal and IL-1β stimulated MMP-1, -3, and -13, possibly through inhibition of NFκB19. However, another in vitro study found that Hedgehog proteins stimulated accumulation of superficial zone protein (lubricin) while antagonists of Wnt/β-catenin also stimulated lubricin accumulation in explants. Indian hedgehog, which can regulate Wnt signaling as well as PTHrp, was found to be increased in human OA cartilage and synovium and promote chondrocyte hypertrophy including increasing MMP-1320. These studies suggest that a much more complete understanding of Wnt signaling pathways in cartilage and related joint tissues will be needed before rational therapeutic interventions can be designed.

Role of inflammatory factors in OA

Studies of pro-inflammatory mediators found in the OA joint support the paradigm that although OA may be largely a biomechanical disease, its manifestations at the tissue level are mediated by the activity of inflammatory factors. In addition, post-traumatic OA is an example of a form of OA where both acute inflammation induced by joint injury and abnormal biomechanics resulting from joint instability likely work in concert to promote joint tissue destruction. Meniscal injury, either alone or in combination with an ACL tear, is an important risk factor for knee OA. A study of meniscal tissue removed during partial meniscectomy in patients with a meniscal tear, with or without a concomitant ACL tear, provided evidence that the injured meniscus can be a source of inflammatory mediators in the joint 21. Numerous cytokines, chemokines, and matrix degrading proteinases were found to be expressed in the meniscus samples including IL-1α and -β, IL-6, TNF-α, CCL3, CXCL1, CXCL3, ADAMTS-4 and -5, MMP-1, MMP-9, and MMP-13.

In a post-traumatic OA model in mice that involves fracture of the lateral tibial plateau, the role of inflammation was studied by measuring inflammatory mediators in the synovial fluid and expression in the synovium in two different strains of mice22. Male MRL/MpJ mice developed less severe OA than C57BL/6 mice despite having a similar joint injury. The C57BL/6 mice were found to have higher levels of IL-1-α and IL–β in the synovial fluid and greater expression of TNF-α and IL-1-β as well as the chemokine CCL22 in synovial tissue where a greater infiltration of macrophages was also found. These finds support the hypothesis that a more active inflammatory response contributed to more severe OA in these mice.

In addition to cytokines and chemokines, many other pro-inflammatory factors have been studied for their potential involvement in OA including the S100 proteins. S100A8 and S100A9 are referred to as alarmins or as damage-associated molecular patterns (DAMPs) since they are released in response to tissue injury and their presence alarms the innate immune system that tissue damage has occurred. Both proteins were found to be present in human synovial tissue removed by biopsy of patients with early symptomatic OA23. Macrophages are one potential source of S100 proteins in the joint and in vitro treatment of macrophages with basic calcium phosphate crystals, which can be involved in OA, stimulated production of S100A824. Two mouse models of OA with different levels of synovial inflammation were compared using S100A9 knockout mice (S100A8 knockouts are not viable and so were not tested). In the less inflammatory DMM model, S100A9 knockout did not protect the articular cartilage while in the collagenase-induced OA model, where much more synovitis was noted, knockout mice had less severe synovitis and cartilage destruction23. In further studies, S100A8 and A9 were identified in human OA cartilage and stimulation of chondrocytes in vitro resulted in increased production of IL-6, IL-8, and MCP-125.

S100A8 and A9 can induce production of pro-inflammatory mediators through Toll-like receptors (TLRs) which are gaining attention for their role in OA. Inhibition of TLR-4 inhibited catabolic effects of S100 A8 and A9 when chondrocytes were stimulated25. Additional pro-inflammatory mediators, some of which also activate TLRs, were identified in human OA synovial fluid using a proteomics approach26. These included plasma proteins such as Gc-globulin, α1-microglobulin, and α2-microglobulin. Of interest, this study also measured the amounts of several cytokines in OA synovial fluids and compared the levels to those in serum. Two widely studied cytokines, IL-1β and TNF-α, were present at very low levels in synovial fluid while IL-6, VEGF, MCP-1, IP-10, and MIG were all present at higher levels with IP-10 and MIG levels over 1000-fold higher than IL-1β26. Another study looked for TLR activators in synovial fluid from patients with early cartilage damage who were undergoing arthroscopy to treat a meniscal injury27. This strategy resulted in the discovery of a TLR co-receptor, soluble CD14, which augmented the ability of TLR-2 and -4 ligands to stimulate IL-8 release from synovial fibroblasts.

In addition to promoting joint tissue destruction, inflammatory mediators may also contribute to pain in people with OA. Using the mouse DMM model of OA, Miller et al showed that the mice exhibit allodynia and discovered that the chemokine MCP-1 (CCL2) and its receptor CCR2 were increased in the dorsal root ganglia28. CCR2 knockouts exhibited similar cartilage damage to wild type mice after DMM surgery but had reduced movement-evoked pain behavior and some resolution of allodynia. A possible role for macrophages in this process was indicated by the finding of macrophages in the dorsal root ganglia of the DMM mice which were reduced in the CCR2 knockouts. These studies support an important role for macrophages and inflammation in OA but suggest that different factors may contribute to macrophage infiltration that contributes to pain and infiltration in the synovium that contributes to joint tissue destruction.

Protective role of lubricin (PGR4) in OA

Changes in lubrication at the surface of articular cartilage in response to injury and altered mechanics have emerged as important in the biology of OA. Much of the recent attention in this area has been on lubricin which is also known as superficial zone protein or as proteoglycan protein 4 (PRG4). When compared to normal synovial fluid, synovial fluid from human OA joints that had reduced lubricin levels was found to have reduced cartilage boundary-lubricating ability when tested in a friction test29. Supplementing these samples with lubricin restored the lubricating ability. Testing done in lubricin knockout mice demonstrated increases in joint friction and chondrocyte apoptosis in the superficial cartilage that appeared to be related30. Loss of lubricin from the articular surface was seen in the rat ACLT model. Supplementation with lubricin by intra-articular injection improved weight bearing in a measurement of hind limb force and reduced the level of urinary CTX-II but did not alter the OARSI score31.

Finally, in a very exciting study32 over-expression of lubricin in transgenic mice reduced the severity of both age-related OA (tested at 10 months of age) as well as in a cruciate ligament transection model of OA. The severity of post-traumatic OA was also reduced by expressing lubricin in the mouse knee joint using intra-articular injection of adeno-associated virus, even when injected 2 weeks after the joint injury. The study went on to examine how lubricin affected gene transcription using superficial chondrocytes for RNA isolation and analysis by microarrays. It was discovered that expression of lubricin inhibited expression of genes involved in cartilage catabolism and chondrocyte hypertrophy in association with an up-regulation of HIF-3α, a negative regulator of HIF-1α and HIF-2α. The authors proposed that the increase in HIF-3α may correct the imbalance in catabolic and anabolic activity that promotes OA through down-regulation of HIF-1α and HIF-2α32. These studies support further investigation of lubricin (PRG4, SZP) as a therapeutic agent for human OA, perhaps given as soon as possible after an acute joint injury to prevent post-traumatic OA.

The role of altered FGF signaling in OA

Unlike the consistent pro-anabolic effects observed with FGF-18, FGF-2 effects in adult articular cartilage have been less clear. Previous in vitro experiments found FGF-2 stimulated chondrocyte proliferation, inhibited matrix synthesis, and promoted MMP-13 production33, 34 ENREF 33. FGF-2 released when cartilage is cut with a scalpel ex vivo (a proposed model of cartilage injury) resulted in activation of MAP kinases and IKK35 that might be expected to promote pro-inflammatory gene expression. However, FGF-2 knockout mice were found to develop accelerated OA with aging and more severe OA in the DMM model36. Experiments showing differences in the response of human and mouse cartilage to FGF-2 suggested that differences in expression of FGF receptors (FGFR)-1 and FGFR-3 might be to blame for the inconsistent findings37. Earlier studies had shown deletion of FGFR-3 in mice resulted in spontaneous OA suggesting FGF-2 activation of FGFR-3 might be chondroprotective38 while FGFR-1 was proposed to mediate catabolic effects of FGF-2 based on in vitro experiments. In support of catabolic activity in response to FGFR-1 activation, Weng et al found that FGFR-1 deficient mice developed less severe age-related aggrecan loss at 12 months and less severe OA in younger mice using the DMM model that was associated with decreased expression of MMP-13 and interestingly higher expression of FGFR-339.

Taken together, these studies suggest that the ratio of FGFR-1 and FGFR-3 in articular cartilage determines the response of chondrocytes to FGF stimulation such that an excess of FGFR-1 stimulation relative to FGFR-3 would promote cartilage catabolism and the development of OA. Previous work had demonstrated the ratio of FGFR-3 to FGFR-1 was reduced in OA40. This is similar to the finding that the ratio of the TGF-β receptors Alk-1 and Alk-5 determine the response to TGF-β stimulation such that an increased Alk-1 to Alk-5 ratio in OA is pro-catabolic. Since FGF-2 can stimulate either FGFR-1 or FGFR-3 and TGF-β can stimulate either Alk-1 or Alk-5, they would be unlikely to be of much benefit in treating OA when the relative receptor levels favor catabolic over anabolic activity. However, FGF-18 primarily stimulates FGFR-3 and so would be expected to promote anabolic over catabolic activity making it a better choice for treating OA. Currently, intra-articular FGF-18 is in early phase studies as an anabolic agent to promote cartilage repair and to treat OA (clinicaltrials.gov)

Bone

The role of bone in the biology of OA has been an active area of investigation. Although OA changes in the articular cartilage in some humans and animal models (see above studies on the DMM model in mice) can be seen in the absence of significant synovitis, involvement of bone proximate to the affected joint seems to be universal during at least some stage of OA. However, various studies reported during the last year that used either human tissue or animal models found both a good correlation between changes occurring in bone and in the overlying articular cartilage or a poor correlation between the two. Human osteochondral plugs taken from tibial plateaus with a range of cartilage damage were found by micro-CT to have changes in subchondral bone mineralization and bone volume underneath the samples with the most severe cartilage changes41. The same group published a second study investigating the contribution of excessive loading and endochondral ossification to the subchondral bone changes seen in OA and concluded that both processes were responsible42.

Studies in various mouse models of OA noted both a good and poor correlation between cartilage and bone changes. A study using the DMM model to compare OA severity in a strain of mice (LGXSM-6) noted to be “super healers” and a strain (LGXSM-33) noted to be “poor healers” of cartilage and ear punch lesions found that the more severe cartilage lesions in the poor healer strain correlated with more severe bone changes measured by micro-CT43. Likewise, bone targeted overexpression of EphB4 resulted in reduced cartilage damage in the DMM model that correlated with a reduction in subchondral sclerosis and bone volume as well as the number of tartrate-resistant acid phosphatase-positive osteoclasts44. In contrast to these studies, compared to wild-type controls, Col6a1-/- mice surprisingly had less knee joint cartilage loss at 15 months of age despite large osteophytes which appeared as early as 2 months of age45. As noted above, sclerostin knockouts did not exhibit age-related OA at 12 or 16 months of age despite a significant increase in subchondral bone mass14. A time course study of the DMM model in 12 week old C57BL/6 mice found significant osteophyte (chondrophyte) formation at 2 weeks after DMM surgery which was a time point when the articular cartilage damage was minimal46. In that study, the size of the medial abaxial osteophytes did not progress from 2 to 16 weeks while the cartilage damage and subchondral bone thickening did progress.

Differences in osteophyte formation and cartilage damage were also noted in studies examining the effect of cathepsin K inhibition on the development of OA. Cathepsin K, a protease produced by osteoclasts and involved in bone resorption, has also been shown to be made by synovial fibroblasts and chondrocytes. A recent study found it can contribute to type II collagen cleavage seen with aging and in OA47. Treatment of rabbits with a chemical inhibitor of cathepsin K (L-235) reduced cartilage damage measured by Mankin scoring in the ACLT model and cathepsin K knockout mice likewise had reduced cartilage damage after ACLT surgery48. In both models, inhibition of cathepsin K protected the subchondral bone. However, a significant reduction in osteophytes was only seen in the rabbit ACLT model where the chemical inhibitor was only partially protective in terms of cartilage damage while in the cathepsin K -/- mice no reduction in osteophytes was seen despite a very significant reduction in the Mankin score.

Conclusions

The various tissues involved in the OA process and a host of potential mediators that contribute to the development of OA continue to be defined (Figure 1). The ultimate goal of research on the biology of OA is to discover new therapeutic targets based on a better understanding of disease mechanisms. Although epigenetic regulation of gene transcription and Wnt signaling both appear to contribute to the OA process, the complexity of the interacting systems involved and the difficulty in targeting one aspect of their activity, which might be beneficial in OA, without affecting another aspect that would be harmful, will make these areas difficult to translate to new therapeutics. It is not to say that they are not important areas for continued research but rather translation to the clinic will require more precisely defined targets and will be much slower than in other areas.

Fig. 1.

Fig. 1

Interactions among tissues and biological mediators that contribute to the biology of OA.

The knowledge of the role of inflammation in OA is rapidly expanding and there is evidence that pro-inflammatory mediators can be produced by all of the joint tissues affected by OA. The limitation in this area of research is that no single target stands out above the others. Unlike in RA, where targeting TNFα results in significant improvement in symptoms and disease progression in many (but not all) patients, it appears that in OA there may be subsets of patients where inflammation is a driving force and within this subset more than one pro-inflammatory mediator may need to be inhibited. It is very possible, as noted by the work on the chemokine receptor CCR2, that pro-inflammatory mediators responsible for pain differ from those responsible for joint tissue destruction.

Closer to translation as new therapeutics for knee OA, intra-articular treatment with lubricin or with anabolic factors, such as FGF-18, is supported by recent work on the biology of OA. The finding that an injection of lubricin given 2 weeks after a joint injury can reduce the severity of subsequent OA in mice suggests that it could be a useful treatment to prevent post-traumatic OA. The ability of FGF-18 to activate FGFR3 and promote anabolic activity in joint tissues holds promise with the caveat that a loss of FGFR3 and gain of FGRF1 in OA cartilage could limit its effectiveness at more advanced stages of disease.

Finally, a review of the most recent literature on bone in OA supports the long-held belief that bone is involved in the OA disease process. However, the association between changes in bone and destruction of the overlying cartilage is not consistent, particularly in regards to osteophyte formation. This suggests that, similar to the role of inflammation, there are likely to be subsets of OA where bone is a driving factor in the disease process and osteophytes are not the best marker to define these subsets. Missing from this review is a discussion of the focal areas of bone remodeling that can be detected by imaging modalities such as MRI and which may be important in pain and/or disease progression. Clearly the OA field is rapidly advancing but a better understanding of the biology of the disease that can translate to new therapeutics will require a division of OA into subsets of disease which will have different etiologies rather than lumping all of OA into a single disease process.

Table 1. Major Topics and Findings in the Biology of OA: 2013.

Topic New Findings Therapeutic Implications References
Epigenetics
  • ↓ methylation of MMP-13, IL-1β, and iNOS promoters.

  • ↑ methylation of Sox-9 promoter.

  • Altered histone acetylation and differential miRNA expression in OA chondrocytes.

Histone deacetylase inhibitors decrease OA in a mouse model of OA. DNA methylation and miRNA expression will be much more difficult to target for therapy. 2-13
Wnt Signaling
  • Sclerostin present in articular cartilage.

  • ↑Dkk-1 in OA cartilage and synovium.

  • ↑WISP-3/CCN6 in OA cartilage.

Sclerostin inhibition/deletion did not affect the development of OA in rat or mouse models. Inconclusive results with Dkk-1 inhibition/overexpression or manipulation of Wnt/β-catenin activity. 14-20
Inflammatory Factors
  • Production of multiple pro-inflammatory mediators by the injured meniscus.

  • S100A8 and S100A9 present in OA synovium.

  • Plasma proteins and soluble CD14 present in SF which activate TLRs.

  • CCL2→CCR2 involved in pain in OA.

  • ↓ OA in S100A8/A9 -/- mice in collagenase-induced OA but not in DMM model.

  • ↓ pain but not OA in CCR2 -/- mice in the DMM model

21-28
Lubricin (PRG4) Important boundary lubricant that protects articular surface. Overexpression of lubricin reduces OA severity in mouse models of OA. 29-32
FGF Signaling Balance of FGFR1 to FGFR3 mediated signaling determines catabolic vs anabolic activity of FGFs.
  • Less severe OA in FGFR1-/- mice.

  • Activation of FGFR3 by FGF-18 more likely to reduce OA than FGFR1 activation by FGF-2.

33-40
Bone Correlation between cartilage and bone changes in OA varies by model system studied, particularly when osteophytes are examined. Subsets of OA where bone is a driving factor appear most likely. Inhibition of cathepsin K ↓OA severity in rabbit and mouse models. 14, 41-48.

Acknowledgments

Dr. Loeser's work has been supported by grants from the NIH (R37 AR049003 and RO1 AG044034) and the Arthritis Foundation as well as the Dorothy Rhyne Kimbrell and Willard Duke Kimbrell Professorship.

Footnotes

Author contribution: Richard F. Loeser searched the literature, summarized the results, and wrote the manuscript.

Conflict of interests: The author has no competing interests.

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.van der Kraan PM. Osteoarthritis year 2012 in review: biology. Osteoarthritis Cartilage. 2012;20:1447–1450. doi: 10.1016/j.joca.2012.07.010. [DOI] [PubMed] [Google Scholar]
  • 2.Bui C, Barter MJ, Scott JL, Xu Y, Galler M, Reynard LN, et al. cAMP response element-binding (CREB) recruitment following a specific CpG demethylation leads to the elevated expression of the matrix metalloproteinase 13 in human articular chondrocytes and osteoarthritis. FASEB J. 2012;26:3000–3011. doi: 10.1096/fj.12-206367. [DOI] [PubMed] [Google Scholar]
  • 3.Hashimoto K, Otero M, Imagawa K, de Andres MC, Coico JM, Roach HI, et al. Regulated transcription of human matrix metalloproteinase 13 (MMP13) and interleukin-1beta (IL1B) genes in chondrocytes depends on methylation of specific proximal promoter CpG Sites. J Biol Chem. 2013;288:10061–10072. doi: 10.1074/jbc.M112.421156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.de Andres MC, Imagawa K, Hashimoto K, Gonzalez A, Roach HI, Goldring MB, et al. Loss of methylation in CpG sites in the NF-kappaB enhancer elements of inducible nitric oxide synthase is responsible for gene induction in human articular chondrocytes. Arthritis Rheum. 2013;65:732–742. doi: 10.1002/art.37806. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Kim KI, Park YS, Im GI. Changes in the epigenetic status of the SOX-9 promoter in human osteoarthritic cartilage. J Bone Miner Res. 2013;28:1050–1060. doi: 10.1002/jbmr.1843. [DOI] [PubMed] [Google Scholar]
  • 6.Saito T, Nishida K, Furumatsu T, Yoshida A, Ozawa M, Ozaki T. Histone deacetylase inhibitors suppress mechanical stress-induced expression of RUNX-2 and ADAMTS-5 through the inhibition of the MAPK signaling pathway in cultured human chondrocytes. Osteoarthritis Cartilage. 2013;21:165–174. doi: 10.1016/j.joca.2012.09.003. [DOI] [PubMed] [Google Scholar]
  • 7.Culley KL, Hui W, Barter MJ, Davidson RK, Swingler TE, Destrument AP, et al. Class I HDAC inhibition modulates metalloproteinase expression and blocks cytokine-induced cartilage degradation. Arthritis Rheum. 2013 doi: 10.1002/art.37965. [DOI] [PubMed] [Google Scholar]
  • 8.Delgado-Calle J, Fernandez AF, Sainz J, Zarrabeitia MT, Sanudo C, Garcia-Renedo R, et al. Genome-wide profiling of bone reveals differentially methylated regions in osteoporosis and osteoarthritis. Arthritis Rheum. 2013;65:197–205. doi: 10.1002/art.37753. [DOI] [PubMed] [Google Scholar]
  • 9.Diaz-Prado S, Cicione C, Muinos-Lopez E, Hermida-Gomez T, Oreiro N, Fernandez-Lopez C, et al. Characterization of microRNA expression profiles in normal and osteoarthritic human chondrocytes. BMC Musculoskelet Disord. 2012;13:144. doi: 10.1186/1471-2474-13-144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Ukai T, Sato M, Akutsu H, Umezawa A, Mochida J. MicroRNA-199a-3p, microRNA-193b, and microRNA-320c are correlated to aging and regulate human cartilage metabolism. J Orthop Res. 2012;30:1915–1922. doi: 10.1002/jor.22157. [DOI] [PubMed] [Google Scholar]
  • 11.Akhtar N, Haqqi TM. MicroRNA-199a* regulates the expression of cyclooxygenase-2 in human chondrocytes. Ann Rheum Dis. 2012;71:1073–1080. doi: 10.1136/annrheumdis-2011-200519. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Liang ZJ, Zhuang H, Wang GX, Li Z, Zhang HT, Yu TQ, et al. MiRNA-140 is a negative feedback regulator of MMP-13 in IL-1beta-stimulated human articular chondrocyte C28/I2 cells. Inflamm Res. 2012;61:503–509. doi: 10.1007/s00011-012-0438-6. [DOI] [PubMed] [Google Scholar]
  • 13.Miyaki S, Sato T, Inoue A, Otsuki S, Ito Y, Yokoyama S, et al. MicroRNA-140 plays dual roles in both cartilage development and homeostasis. Genes Dev. 2010;24:1173–1185. doi: 10.1101/gad.1915510. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Roudier M, Li X, Niu QT, Pacheco E, Pretorius J, Graham K, et al. Sclerostin is expressed in articular cartilage but loss or inhibition does not affect cartilage remodeling during aging or following mechanical injury. Arthritis Rheum. 2012;65:721–731. doi: 10.1002/art.37802. [DOI] [PubMed] [Google Scholar]
  • 15.Oh H, Chun CH, Chun JS. Dkk-1 expression in chondrocytes inhibits experimental osteoarthritic cartilage destruction in mice. Arthritis Rheum. 2012;64:2568–2578. doi: 10.1002/art.34481. [DOI] [PubMed] [Google Scholar]
  • 16.Weng LH, Ko JY, Wang CJ, Sun YC, Wang FS. Dkk-1 promotes angiogenic responses and cartilage matrix proteinase secretion in synovial fibroblasts from osteoarthritic joints. Arthritis Rheum. 2012;64:3267–3277. doi: 10.1002/art.34602. [DOI] [PubMed] [Google Scholar]
  • 17.Leijten JC, Emons J, Sticht C, van Gool S, Decker E, Uitterlinden A, et al. Gremlin 1, frizzled-related protein, and Dkk-1 are key regulators of human articular cartilage homeostasis. Arthritis Rheum. 2012;64:3302–3312. doi: 10.1002/art.34535. [DOI] [PubMed] [Google Scholar]
  • 18.Baker N, Sharpe P, Culley K, Otero M, Bevan D, Newham P, et al. Dual regulation of metalloproteinase expression in chondrocytes by Wnt-1-inducible signaling pathway protein 3/CCN6. Arthritis Rheum. 2012;64:2289–2299. doi: 10.1002/art.34411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Ma B, van Blitterswijk CA, Karperien M. A Wnt/beta-catenin negative feedback loop inhibits interleukin-1-induced matrix metalloproteinase expression in human articular chondrocytes. Arthritis Rheum. 2012;64:2589–2600. doi: 10.1002/art.34425. [DOI] [PubMed] [Google Scholar]
  • 20.Wei F, Zhou J, Wei X, Zhang J, Fleming BC, Terek R, et al. Activation of Indian hedgehog promotes chondrocyte hypertrophy and upregulation of MMP-13 in human osteoarthritic cartilage. Osteoarthritis Cartilage. 2012;20:755–763. doi: 10.1016/j.joca.2012.03.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Brophy RH, Rai MF, Zhang Z, Torgomyan A, Sandell LJ. Molecular analysis of age and sex-related gene expression in meniscal tears with and without a concomitant anterior cruciate ligament tear. J Bone Joint Surg Am. 2012;94:385–393. doi: 10.2106/JBJS.K.00919. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Lewis JS, Jr, Furman BD, Zeitler E, Huebner JL, Kraus VB, Guilak F, et al. Genetic and cellular evidence of decreased inflammation associated with reduced incidence of posttraumatic arthritis in MRL/MpJ mice. Arthritis Rheum. 2013;65:660–670. doi: 10.1002/art.37796. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.van Lent PL, Blom AB, Schelbergen RF, Sloetjes A, Lafeber FP, Lems WF, et al. Active involvement of alarmins S100A8 and S100A9 in the regulation of synovial activation and joint destruction during mouse and human osteoarthritis. Arthritis Rheum. 2012;64:1466–1476. doi: 10.1002/art.34315. [DOI] [PubMed] [Google Scholar]
  • 24.Cunningham CC, Mills E, Mielke LA, O'Farrell LK, Lavelle E, Mori A, et al. Osteoarthritis-associated basic calcium phosphate crystals induce pro-inflammatory cytokines and damage-associated molecules via activation of Syk and PI3 kinase. Clin Immunol. 2012;144:228–236. doi: 10.1016/j.clim.2012.06.007. [DOI] [PubMed] [Google Scholar]
  • 25.Schelbergen RF, Blom AB, van den Bosch MH, Sloetjes A, Abdollahi-Roodsaz S, Schreurs BW, et al. Alarmins S100A8 and S100A9 elicit a catabolic effect in human osteoarthritic chondrocytes that is dependent on Toll-like receptor 4. Arthritis Rheum. 2012;64:1477–1487. doi: 10.1002/art.33495. [DOI] [PubMed] [Google Scholar]
  • 26.Sohn DH, Sokolove J, Sharpe O, Erhart JC, Chandra PE, Lahey LJ, et al. Plasma proteins present in osteoarthritic synovial fluid can stimulate cytokine production via Toll-like receptor 4. Arthritis Res Ther. 2012;14:R7. doi: 10.1186/ar3555. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Nair A, Kanda V, Bush-Joseph C, Verma N, Chubinskaya S, Mikecz K, et al. Synovial fluid from patients with early osteoarthritis modulates fibroblast-like synoviocyte responses to toll-like receptor 4 and toll-like receptor 2 ligands via soluble CD14. Arthritis Rheum. 2012;64:2268–2277. doi: 10.1002/art.34495. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Miller RE, Tran PB, Das R, Ghoreishi-Haack N, Ren D, Miller RJ, et al. CCR2 chemokine receptor signaling mediates pain in experimental osteoarthritis. Proc Natl Acad Sci U S A. 2012;109:20602–20607. doi: 10.1073/pnas.1209294110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Ludwig TE, McAllister JR, Lun V, Wiley JP, Schmidt TA. Diminished cartilage-lubricating ability of human osteoarthritic synovial fluid deficient in proteoglycan 4: Restoration through proteoglycan 4 supplementation. Arthritis Rheum. 2012;64:3963–3971. doi: 10.1002/art.34674. [DOI] [PubMed] [Google Scholar]
  • 30.Waller KA, Zhang LX, Elsaid KA, Fleming BC, Warman ML, Jay GD. Role of lubricin and boundary lubrication in the prevention of chondrocyte apoptosis. Proc Natl Acad Sci U S A. 2013;110:5852–5857. doi: 10.1073/pnas.1219289110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Jay GD, Elsaid KA, Kelly KA, Anderson SC, Zhang L, Teeple E, et al. Prevention of cartilage degeneration and gait asymmetry by lubricin tribosupplementation in the rat following anterior cruciate ligament transection. Arthritis Rheum. 2012;64:1162–1171. doi: 10.1002/art.33461. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Ruan MZ, Erez A, Guse K, Dawson B, Bertin T, Chen Y, et al. Proteoglycan 4 expression protects against the development of osteoarthritis. Sci Transl Med. 2013;5:176ra134. doi: 10.1126/scitranslmed.3005409. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Loeser RF, Chubinskaya S, Pacione C, Im HJ. 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. doi: 10.1002/art.21472. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Im HJ, Muddasani P, Natarajan V, Schmid TM, Block JA, Davis F, et al. 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. doi: 10.1074/jbc.M609040200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Watt FE, Ismail HM, Didangelos A, Peirce M, Vincent TL, Wait R, et al. Src and fibroblast growth factor 2 independently regulate signaling and gene expression induced by experimental injury to intact articular cartilage. Arthritis Rheum. 2013;65:397–407. doi: 10.1002/art.37765. [DOI] [PubMed] [Google Scholar]
  • 36.Chia SL, Sawaji Y, Burleigh A, McLean C, Inglis J, Saklatvala J, et al. 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. doi: 10.1002/art.24654. [DOI] [PubMed] [Google Scholar]
  • 37.Li X, Ellman MB, Kroin JS, Chen D, Yan D, Mikecz K, et al. Species-specific biological effects of FGF-2 in articular cartilage: implication for distinct roles within the FGF receptor family. J Cell Biochem. 2012;113:2532–2542. doi: 10.1002/jcb.24129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Valverde-Franco G, Binette JS, Li W, Wang H, Chai S, Laflamme F, et al. Defects in articular cartilage metabolism and early arthritis in fibroblast growth factor receptor 3 deficient mice. Hum Mol Genet. 2006;15:1783–1792. doi: 10.1093/hmg/ddl100. [DOI] [PubMed] [Google Scholar]
  • 39.Weng T, Yi L, Huang J, Luo F, Wen X, Du X, et al. Genetic inhibition of fibroblast growth factor receptor 1 in knee cartilage attenuates the degeneration of articular cartilage in adult mice. Arthritis Rheum. 2012;64:3982–3992. doi: 10.1002/art.34645. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Yan D, Chen D, Cool SM, van Wijnen AJ, Mikecz K, Murphy G, et al. Fibroblast growth factor receptor 1 is principally responsible for fibroblast growth factor 2-induced catabolic activities in human articular chondrocytes. Arthritis Res Ther. 2011;13:R130. doi: 10.1186/ar3441. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 41.Cox LG, van Donkelaar CC, van Rietbergen B, Emans PJ, Ito K. Decreased bone tissue mineralization can partly explain subchondral sclerosis observed in osteoarthritis. Bone. 2012;50:1152–1161. doi: 10.1016/j.bone.2012.01.024. [DOI] [PubMed] [Google Scholar]
  • 42.Cox LG, van Donkelaar CC, van Rietbergen B, Emans PJ, Ito K. Alterations to the subchondral bone architecture during osteoarthritis: bone adaptation vs endochondral bone formation. Osteoarthritis Cartilage. 2013;21:331–338. doi: 10.1016/j.joca.2012.10.013. [DOI] [PubMed] [Google Scholar]
  • 43.Hashimoto S, Rai MF, Janiszak KL, Cheverud JM, Sandell LJ. Cartilage and bone changes during development of post-traumatic osteoarthritis in selected LGXSM recombinant inbred mice. Osteoarthritis Cartilage. 2012;20:562–571. doi: 10.1016/j.joca.2012.01.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Valverde-Franco G, Pelletier JP, Fahmi H, Hum D, Matsuo K, Lussier B, et al. In vivo bone-specific EphB4 overexpression in mice protects both subchondral bone and cartilage during osteoarthritis. Arthritis Rheum. 2012;64:3614–3625. doi: 10.1002/art.34638. [DOI] [PubMed] [Google Scholar]
  • 45.Christensen SE, Coles JM, Zelenski NA, Furman BD, Leddy HA, Zauscher S, et al. Altered trabecular bone structure and delayed cartilage degeneration in the knees of collagen VI null mice. PLoS One. 2012;7:e33397. doi: 10.1371/journal.pone.0033397. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Loeser RF, Olex AL, McNulty MA, Carlson CS, Callahan M, Ferguson C, et al. Disease progression and phasic changes in gene expression in a mouse model of osteoarthritis. PLoS One. 2013;8:e54633. doi: 10.1371/journal.pone.0054633. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Dejica VM, Mort JS, Laverty S, Antoniou J, Zukor DJ, Tanzer M, et al. Increased type II collagen cleavage by cathepsin K and collagenase activities with aging and osteoarthritis in human articular cartilage. Arthritis Res Ther. 2012;14:R113. doi: 10.1186/ar3839. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Hayami T, Zhuo Y, Wesolowski GA, Pickarski M, Duong le T. Inhibition of cathepsin K reduces cartilage degeneration in the anterior cruciate ligament transection rabbit and murine models of osteoarthritis. Bone. 2012;50:1250–1259. doi: 10.1016/j.bone.2012.03.025. [DOI] [PubMed] [Google Scholar]

RESOURCES