Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

Role of proinflammatory cytokines in the pathophysiology of osteoarthritis

Abstract

Osteoarthritis (OA) is associated with cartilage destruction, subchondral bone remodeling and inflammation of the synovial membrane, although the etiology and pathogenesis underlying this debilitating disease are poorly understood. Secreted inflammatory molecules, such as proinflammatory cytokines, are among the critical mediators of the disturbed processes implicated in OA pathophysiology. Interleukin (IL)-1β and tumor necrosis factor (TNF), in particular, control the degeneration of articular cartilage matrix, which makes them prime targets for therapeutic strategies. Animal studies provide support for this approach, although only a few clinical studies have investigated the efficacy of blocking these proinflammatory cytokines in the treatment of OA. Apart from IL-1β and TNF, several other cytokines including IL-6, IL-15, IL-17, IL-18, IL-21, leukemia inhibitory factor and IL-8 (a chemokine) have also been shown to be implicated in OA and could possibly be targeted therapeutically. This Review discusses the current knowledge regarding the role of proinflammatory cytokines in the pathophysiology of OA and addresses the potential of anticytokine therapy in the treatment of this disease.

Key Points

  • Osteoarthritis (OA) is associated with cartilage destruction, subchondral bone remodeling and synovial membrane inflammation

  • Proinflammatory cytokines are critical mediators in the disturbed metabolism and enhanced catabolism of tissue in the OA joint

  • Interleukin (IL)-1β, tumor necrosis factor (TNF) and IL-6 seem to be the main proinflammatory cytokines involved in the pathophysiology of OA

  • Data from cellular and animal studies have provided substantial evidence that blocking IL-1β and TNF production could counteract the degradative mechanisms associated with OA pathology

  • Anticytokine therapies in OA clinical trials have so far yielded variable results

  • A better understanding of the individual roles and functions of cytokines, such as IL-1β, TNF and IL-6, is of the utmost importance in order to develop adequate and specific anticytokine therapies

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: The role of proinflammatory cytokines in the pathophysiology of OA.

References

  1. Felson, D. T. Clinical practice. Osteoarthritis of the knee. N. Engl. J. Med. 354, 841–848 (2006).

    Article  CAS  PubMed  Google Scholar 

  2. Ayral, X., Pickering, E. H., Woodworth, T. G., Mackillop, N. & Dougados, M. Synovitis: a potential predictive factor of structural progression of medial tibiofemoral knee osteoarthritis—results of a 1 year longitudinal arthroscopic study in 422 patients. Osteoarthritis Cartilage 13, 361–367 (2005).

    Article  CAS  PubMed  Google Scholar 

  3. Pozgan, U. et al. Expression and activity profiling of selected cysteine cathepsins and matrix metalloproteinases in synovial fluids from patients with rheumatoid arthritis and osteoarthritis. Biol. Chem. 391, 571–579 (2010).

    Article  CAS  PubMed  Google Scholar 

  4. Martel-Pelletier, J., Lajeunesse, D. & Pelletier, J. P. in Arthritis and Allied Conditions. A Textbook of Rheumatology 15th edn (eds Koopman, W. J. & Moreland, L. W.) 2199–2226 (Lippincott Williams & Wilkins, Baltimore, USA, 2005).

    Google Scholar 

  5. Page Thomas, D. P., King, B., Stephens, T. & Dingle, J. T. In vivo studies of cartilage regeneration after damage induced by catabolin/interleukin-1. Ann. Rheum. Dis. 50, 75–80 (1991).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Henderson, B. & Pettipher, E. R. Arthritogenic actions of recombinant IL-1 and tumour necrosis factor α in the rabbit: evidence for synergistic interactions between cytokines in vivo. Clin. Exp. Immunol. 75, 306–310 (1989).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Clements, K. M. et al. Gene deletion of either interleukin-1β, interleukin-1β-converting enzyme, inducible nitric oxide synthase, or stromelysin 1 accelerates the development of knee osteoarthritis in mice after surgical transection of the medial collateral ligament and partial medial meniscectomy. Arthritis Rheum. 48, 3452–3463 (2003).

    Article  CAS  PubMed  Google Scholar 

  8. Arend, W. P. Interleukin-1 receptor antagonist. Adv. Immunol. 54, 167–227 (1993).

    Article  CAS  PubMed  Google Scholar 

  9. Martel-Pelletier, J. et al. The interleukin-1 receptor in normal and osteoarthritic human articular chondrocytes. Identification as the type I receptor and analysis of binding kinetics and biologic function. Arthritis Rheum. 35, 530–540 (1992).

    Article  CAS  PubMed  Google Scholar 

  10. Sadouk, M. B. et al. Human synovial fibroblasts coexpress IL-1 receptor type I and type II mRNA. The increased level of the IL-1 receptor in osteoarthritic cells is related to an increased level of the type I receptor. Lab. Invest. 73, 347–355 (1995).

    CAS  PubMed  Google Scholar 

  11. Colotta, F., Dower, S. K., Sims, J. E. & Mantovani, A. The type II 'decoy' receptor: a novel regulatory pathway for interleukin 1. Immunol. Today 15, 562–566 (1994).

    Article  CAS  PubMed  Google Scholar 

  12. Palmer, G. et al. Production of interleukin-1 receptor antagonist by human articular chondrocytes. Arthritis Res. 4, 226–231 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Seitz, M. et al. Production of interleukin-1 receptor antagonist, inflammatory chemotactic proteins, and prostaglandin E by rheumatoid and osteoarthritic synoviocytes—regulation by IFN-γ and IL-4. J. Immunol. 152, 2060–2065 (1994).

    CAS  PubMed  Google Scholar 

  14. Alaaeddine, N. et al. Osteoarthritic synovial fibroblasts possess an increased level of tumor necrosis factor-receptor 55 (TNF-R55) that mediates biological activation by TNFα. J. Rheumatol. 24, 1985–1994 (1997).

    CAS  PubMed  Google Scholar 

  15. Naume, B., Shalaby, R., Lesslauer, W. & Espevik, T. Involvement of the 55- and 75-kDa tumor necrosis factor receptors in the generation of lymphokine-activated killer cell activity and proliferation of natural killer cells. J. Immunol. 146, 3045–3048 (1991).

    CAS  PubMed  Google Scholar 

  16. Hohmann, H. P. et al. Expression of the types A and B tumor necrosis factor (TNF) receptors is independently regulated, and both receptors mediate activation of the transcription factor NF-κB. TNFα is not needed for induction of a biological effect via TNF receptors. J. Biol. Chem. 265, 22409–22417 (1990).

    CAS  PubMed  Google Scholar 

  17. Westacott, C. I., Atkins, R. M., Dieppe, P. A. & Elson, C. J. Tumor necrosis factor α receptor expression on chondrocytes isolated from human articular cartilage. J. Rheumatol. 21, 1710–1715 (1994).

    CAS  PubMed  Google Scholar 

  18. Saklatvala, J. Tumour necrosis factor α stimulates resorption and inhibits synthesis of proteoglycan in cartilage. Nature 322, 547–549 (1986).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Goldring, M. B., Fukuo, K., Birkhead, J. R., Dudek, E. & Sandell, L. J. Transcriptional suppression by interleukin-1 and interferon-γ of type II collagen gene expression in human chondrocytes. J. Cell. Biochem. 54, 85–99 (1994).

    Article  CAS  PubMed  Google Scholar 

  20. Chadjichristos, C. et al. Sp1 and Sp3 transcription factors mediate interleukin-1β down-regulation of human type II collagen gene expression in articular chondrocytes. J. Biol. Chem. 278, 39762–39772 (2003).

    Article  CAS  PubMed  Google Scholar 

  21. Shakibaei, M., Schulze-Tanzil, G., John, T. & Mobasheri, A. Curcumin protects human chondrocytes from IL-l1β -induced inhibition of collagen type II and β1-integrin expression and activation of caspase-3: an immunomorphological study. Ann. Anat. 187, 487–497 (2005).

    Article  CAS  PubMed  Google Scholar 

  22. Stöve, J., Huch, K., Günther, K. P. & Scharf, H. P. Interleukin-1β induces different gene expression of stromelysin, aggrecan and tumor-necrosis-factor-stimulated gene 6 in human osteoarthritic chondrocytes in vitro. Pathobiology 68, 144–149 (2000).

    Article  PubMed  Google Scholar 

  23. Nietfeld, J. J., Wilbrink, B., Den Otter, W., Huber, J. & Huber-Bruning, O. The effect of human interleukin 1 on proteoglycan metabolism in human and porcine cartilage explants. J. Rheumatol. 17, 818–826 (1990).

    CAS  PubMed  Google Scholar 

  24. Gouze, J. N. et al. Interleukin-1β down-regulates the expression of glucuronosyltransferase I, a key enzyme priming glycosaminoglycan biosynthesis: influence of glucosamine on interleukin-1β-mediated effects in rat chondrocytes. Arthritis Rheum. 44, 351–360 (2001).

    Article  CAS  PubMed  Google Scholar 

  25. Séguin, C. A. & Bernier, S. M. TNFα suppresses link protein and type II collagen expression in chondrocytes: Role of MEK1/2 and NF-κB signaling pathways. J. Cell. Physiol. 197, 356–369 (2003).

    Article  CAS  PubMed  Google Scholar 

  26. Mengshol, J. A., Vincenti, M. P., Coon, C. I., Barchowsky, A. & Brinckerhoff, C. E. Interleukin-1 induction of collagenase 3 (matrix metalloproteinase 13) gene expression in chondrocytes requires p38, c-Jun N-terminal kinase, and nuclear factor κB: differential regulation of collagenase 1 and collagenase 3. Arthritis Rheum. 43, 801–811 (2000).

    Article  CAS  PubMed  Google Scholar 

  27. Lefebvre, V., Peeters-Joris, C. & Vaes, G. Modulation by interleukin 1 and tumor necrosis factor α of production of collagenase, tissue inhibitor of metalloproteinases and collagen types in differentiated and dedifferentiated articular chondrocytes. Biochim. Biophys. Acta 1052, 366–378 (1990).

    Article  CAS  PubMed  Google Scholar 

  28. Reboul, P., Pelletier, J. P., Tardif, G., Cloutier, J. M. & Martel-Pelletier, J. The new collagenase, collagenase-3, is expressed and synthesized by human chondrocytes but not by synoviocytes. A role in osteoarthritis. J. Clin. Invest. 97, 2011–2019 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Glasson, S. S. et al. Characterization of and osteoarthritis susceptibility in ADAMTS-4-knockout mice. Arthritis Rheum. 50, 2547–2558 (2004).

    Article  CAS  PubMed  Google Scholar 

  30. Glasson, S. S. et al. Deletion of active ADAMTS5 prevents cartilage degradation in a murine model of osteoarthritis. Nature 434, 644–648 (2005).

    Article  CAS  PubMed  Google Scholar 

  31. Tortorella, M. D., Malfait, A. M., Deccico, C. & Arner, E. The role of ADAM-TS4 (aggrecanase-1) and ADAM-TS5 (aggrecanase-2) in a model of cartilage degradation. Osteoarthritis Cartilage 9, 539–552 (2001).

    Article  CAS  PubMed  Google Scholar 

  32. Bondeson, J., Wainwright, S. D., Lauder, S., Amos, N. & Hughes, C. E. The role of synovial macrophages and macrophage-produced cytokines in driving aggrecanases, matrix metalloproteinases, and other destructive and inflammatory responses in osteoarthritis. Arthritis Res. Ther. 8, R187 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Guerne, P. A., Carson, D. A. & Lotz, M. IL-6 production by human articular chondrocytes. Modulation of its synthesis by cytokines, growth factors, and hormones in vitro. J. Immunol. 144, 499–505 (1990).

    CAS  PubMed  Google Scholar 

  34. Lotz, M., Terkeltaub, R. & Villiger, P. M. Cartilage and joint inflammation. Regulation of IL-8 expression by human articular chondrocytes. J. Immunol. 148, 466–473 (1992).

    CAS  PubMed  Google Scholar 

  35. Villiger, P. M., Terkeltaub, R. & Lotz, M. Monocyte chemoattractant protein-1 (MCP-1) expression in human articular cartilage. Induction by peptide regulatory factors and differential effects of dexamethasone and retinoic acid. J. Clin. Invest. 90, 488–496 (1992).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Alaaeddine, N., Olee, T., Hashimoto, S., Creighton-Achermann, L. & Lotz, M. Production of the chemokine RANTES by articular chondrocytes and role in cartilage degradation. Arthritis Rheum. 44, 1633–1643 (2001).

    Article  CAS  PubMed  Google Scholar 

  37. Afonso, V., Champy, R., Mitrovic, D., Collin, P. & Lomri, A. Reactive oxygen species and superoxide dismutases: role in joint diseases. Joint Bone Spine 74, 324–329 (2007).

    Article  CAS  PubMed  Google Scholar 

  38. Mathy-Hartert, M. et al. Interleukin-1β and interleukin-6 disturb the antioxidant enzyme system in bovine chondrocytes: a possible explanation for oxidative stress generation. Osteoarthritis Cartilage 16, 756–763 (2008).

    Article  CAS  PubMed  Google Scholar 

  39. Scott, J. L. et al. Superoxide dismutase downregulation in osteoarthritis progression and end-stage disease. Ann. Rheum. Dis. 69, 1502–1510 (2010).

    Article  CAS  PubMed  Google Scholar 

  40. Roman-Blas, J. A. & Jimenez, S. A. NF-κB as a potential therapeutic target in osteoarthritis and rheumatoid arthritis. Osteoarthritis Cartilage 14, 839–848 (2006).

    Article  CAS  PubMed  Google Scholar 

  41. Chen, L. X. et al. Suppression of early experimental osteoarthritis by in vivo delivery of the adenoviral vector-mediated NF-κBp65-specific siRNA. Osteoarthritis Cartilage 16, 174–184 (2008).

    Article  CAS  PubMed  Google Scholar 

  42. Blom, A. B. et al. Involvement of the Wnt signaling pathway in experimental and human osteoarthritis: prominent role of Wnt-induced signaling protein 1. Arthritis Rheum. 60, 501–512 (2009).

    Article  CAS  PubMed  Google Scholar 

  43. Yuasa, T., Otani, T., Koike, T., Iwamoto, M. & Enomoto-Iwamoto, M. Wnt/β-catenin signaling stimulates matrix catabolic genes and activity in articular chondrocytes: its possible role in joint degeneration. Lab. Invest. 88, 264–274 (2008).

    Article  CAS  PubMed  Google Scholar 

  44. Lane, N. E., Nevitt, M. C., Lui, L. Y., de Leon, P. & Corr, M. Wnt signaling antagonists are potential prognostic biomarkers for the progression of radiographic hip osteoarthritis in elderly Caucasian women. Arthritis Rheum. 56, 3319–3325 (2007).

    Article  CAS  PubMed  Google Scholar 

  45. Fahmi, H., Pelletier, J. P. & Martel-Pelletier, J. PPARg ligands as modulators of inflammatory and catabolic responses in arthritis. An overview. J. Rheumatol. 29, 3–14 (2002).

    CAS  PubMed  Google Scholar 

  46. Kobayashi, T. et al. Pioglitazone, a peroxisome proliferator-activated receptor γ agonist, reduces the progression of experimental osteoarthritis in guinea pigs. Arthritis Rheum. 52, 479–487 (2005).

    Article  CAS  PubMed  Google Scholar 

  47. Boileau, C. et al. The peroxisome proliferator-activated receptor γ agonist pioglitazone reduces the development of cartilage lesions in an experimental dog model of osteoarthritis: in vivo protective effects mediated through the inhibition of key signaling and catabolic pathways. Arthritis Rheum. 56, 2288–2298 (2007).

    Article  CAS  PubMed  Google Scholar 

  48. Kobayashi, M. et al. Role of interleukin-1 and tumor necrosis factor α in matrix degradation of human osteoarthritic cartilage. Arthritis Rheum. 52, 128–135 (2005).

    Article  CAS  PubMed  Google Scholar 

  49. Caron, J. P. et al. Chondroprotective effect of intraarticular injections of interleukin-1 receptor antagonist in experimental osteoarthritis. Suppression of collagenase-1 expression. Arthritis Rheum. 39, 1535–1544 (1996).

    Article  CAS  PubMed  Google Scholar 

  50. Pelletier, J. P. et al. In vivo suppression of early experimental osteoarthritis by interleukin-1 receptor antagonist using gene therapy. Arthritis Rheum. 40, 1012–1019 (1997).

    Article  CAS  PubMed  Google Scholar 

  51. Fernandes, J. et al . In vivo transfer of interleukin-1 receptor antagonist gene in osteoarthritic rabbit knee joints: prevention of osteoarthritis progression. Am. J. Pathol. 154, 1159–1169 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Zhang, X., Mao, Z. & Yu, C. Suppression of early experimental osteoarthritis by gene transfer of interleukin-1 receptor antagonist and interleukin-10. J. Orthop. Res. 22, 742–750 (2004).

    Article  CAS  PubMed  Google Scholar 

  53. Frisbie, D. D., Ghivizzani, S. C., Robbins, P. D., Evans, C. H. & McIlwraith, C. W. Treatment of experimental equine osteoarthritis by in vivo delivery of the equine interleukin-1 receptor antagonist gene. Gene Ther. 9, 12–20 (2002).

    Article  CAS  PubMed  Google Scholar 

  54. Chevalier, X. et al. Safety study of intraarticular injection of interleukin 1 receptor antagonist in patients with painful knee osteoarthritis: a multicenter study. J. Rheumatol. 32, 1317–1323 (2005).

    CAS  PubMed  Google Scholar 

  55. Chevalier, X. et al. Intraarticular injection of anakinra in osteoarthritis of the knee: a multicenter, randomized, double-blind, placebo-controlled study. Arthritis Rheum. 61, 344–352 (2009).

    Article  CAS  PubMed  Google Scholar 

  56. Bacconnier, L., Jorgensen, C. & Fabre, S. Erosive osteoarthritis of the hand: clinical experience with anakinra. Ann. Rheum. Dis. 68, 1078–1079 (2009).

    Article  CAS  PubMed  Google Scholar 

  57. Attur, M. G. et al. Functional genomic analysis of type II IL-1β decoy receptor: potential for gene therapy in human arthritis and inflammation. J. Immunol. 168, 2001–2010 (2002).

    Article  CAS  PubMed  Google Scholar 

  58. Grunke, M. & Schulze-Koops, H. Successful treatment of inflammatory knee osteoarthritis with tumour necrosis factor blockade. Ann. Rheum. Dis. 65, 555–556 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Fioravanti, A., Fabbroni, M., Cerase, A. & Galeazzi, M. Treatment of erosive osteoarthritis of the hands by intra-articular infliximab injections: a pilot study. Rheumatol. Int. 29, 961–965 (2009).

    Article  CAS  PubMed  Google Scholar 

  60. Magnano, M. D. et al. A pilot study of tumor necrosis factor inhibition in erosive/inflammatory osteoarthritis of the hands. J. Rheumatol. 34, 1323–1327 (2007).

    CAS  PubMed  Google Scholar 

  61. Efficacy study of an anti-tumor necrosis factor (TNF)-α agent in patients with hand osteoarthritis (DORA). http://clinicaltrials.gov [online], (2010).

  62. Skiles, J. W., Gonnella, N. C. & Jeng, A. Y. The design, structure, and clinical update of small molecular weight matrix metalloproteinase inhibitors. Curr. Med. Chem. 11, 2911–2977 (2004).

    Article  CAS  PubMed  Google Scholar 

  63. Wojtowicz-Praga, S. et al. Phase I trial of Marimastat, a novel matrix metalloproteinase inhibitor, administered orally to patients with advanced lung cancer. J. Clin. Oncol. 16, 2150–2156 (1998).

    Article  CAS  PubMed  Google Scholar 

  64. Murphy, G. & Nagase, H. Reappraising metalloproteinases in rheumatoid arthritis and osteoarthritis: destruction or repair? Nat. Clin. Pract. Rheumatol. 4, 128–135 (2008).

    Article  CAS  PubMed  Google Scholar 

  65. Krzeski, P. et al. Development of musculoskeletal toxicity without clear benefit after administration of PG-116800, a matrix metalloproteinase inhibitor, to patients with knee osteoarthritis: a randomized, 12-month, double-blind, placebo-controlled study. Arthritis Res. Ther. 9, R109 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Clutterbuck, A. L., Asplin, K. E., Harris, P., Allaway, D. & Mobasheri, A. Targeting matrix metalloproteinases in inflammatory conditions. Curr. Drug Targets 10, 1245–1254 (2009).

    Article  CAS  PubMed  Google Scholar 

  67. Johnson, A. R. et al. Discovery and characterization of a novel inhibitor of matrix metalloprotease-13 that reduces cartilage damage in vivo without joint fibroplasia side effects. J. Biol. Chem. 282, 27781–27791 (2007).

    Article  CAS  PubMed  Google Scholar 

  68. Tortorella, M. D., Malfait, F., Barve, R. A., Shieh, H. S. & Malfait, A. M. A review of the ADAMTS family, pharmaceutical targets of the future. Curr. Pharm. Des. 15, 2359–2374 (2009).

    Article  CAS  PubMed  Google Scholar 

  69. Marcu, K. B., Otero, M., Olivotto, E., Borzi, R. M. & Goldring, M. B. NF-κB signaling: multiple angles to target OA. Curr. Drug Targets 11, 599–613 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Wegenka, U. M., Buschmann, J., Lütticken, C., Heinrich, P. C. & Horn, F. Acute-phase response factor, a nuclear factor binding to acute-phase response elements, is rapidly activated by interleukin-6 at the posttranslational level. Mol. Cell. Biol. 13, 276–288 (1993).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Zhong, Z., Wen, Z. & Darnell, J. E. Jr. Stat3: a STAT family member activated by tyrosine phosphorylation in response to epidermal growth factor and interleukin-6. Science 264, 95–98 (1994).

    Article  CAS  PubMed  Google Scholar 

  72. Bender, S. et al. Interleukin-1β induces synthesis and secretion of interleukin-6 in human chondrocytes. FEBS Lett. 263, 321–324 (1990).

    Article  CAS  PubMed  Google Scholar 

  73. Wang, P., Zhu, F. & Konstantopoulos, K. Prostaglandin E2 induces interleukin-6 expression in human chondrocytes via cAMP/protein kinase A- and phosphatidylinositol 3-kinase-dependent NF-κB activation. Am. J. Physiol. Cell Physiol. 298, C1445–C1456 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Wang, P., Zhu, F., Lee, N. H. & Konstantopoulos, K. Shear-induced interleukin-6 synthesis in chondrocytes: roles of E prostanoid (EP) 2 and EP3 in cAMP/protein kinase A- and PI3-K/Akt-dependent NF-κB activation. J. Biol. Chem. 285, 24793–24804 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Steensberg, A., Fischer, C. P., Keller, C., Møller, K. & Pedersen, B. K. IL-6 enhances plasma IL-1ra, IL-10, and cortisol in humans. Am. J. Physiol. Endocrinol. Metab. 285, E433–E437 (2003).

    Article  CAS  PubMed  Google Scholar 

  76. Haider, D. G. et al. C-reactive protein is expressed and secreted by peripheral blood mononuclear cells. Clin. Exp. Immunol. 146, 533–539 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Pearle, A. D. et al. Elevated high-sensitivity C-reactive protein levels are associated with local inflammatory findings in patients with osteoarthritis. Osteoarthritis Cartilage 15, 516–523 (2007).

    Article  CAS  PubMed  Google Scholar 

  78. Pelletier, J. P. et al. Decrease in serum level of matrix metalloproteinases is predictive of the disease-modifying effect of osteoarthritis drugs assessed by quantitative MRI in patients with knee osteoarthritis. Ann. Rheum. Dis. doi:10.1136/ard.2009.122002.

  79. Kugisaki, H. et al. Serum concentrations of interleukin-6 in patients following unilateral versus bilateral total knee arthroplasty. J. Orthop. Sci. 14, 437–442 (2009).

    Article  CAS  PubMed  Google Scholar 

  80. Livshits, G. et al. Interleukin-6 is a significant predictor of radiographic knee osteoarthritis: The Chingford Study. Arthritis Rheum. 60, 2037–2045 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Kaneko, S. et al. Interleukin-6 and interleukin-8 levels in serum and synovial fluid of patients with osteoarthritis. Cytokines Cell. Mol. Ther. 6, 71–79 (2000).

    Article  CAS  PubMed  Google Scholar 

  82. Cawston, T. E. et al. The role of oncostatin M in animal and human connective tissue collagen turnover and its localization within the rheumatoid joint. Arthritis Rheum. 41, 1760–1771 (1998).

    Article  CAS  PubMed  Google Scholar 

  83. Rowan, A. D. et al. Synergistic effects of glycoprotein 130 binding cytokines in combination with interleukin-1 on cartilage collagen breakdown. Arthritis Rheum. 44, 1620–1632 (2001).

    Article  CAS  PubMed  Google Scholar 

  84. Sui, Y. et al. Mechanical injury potentiates proteoglycan catabolism induced by interleukin-6 with soluble interleukin-6 receptor and tumor necrosis factor α in immature bovine and adult human articular cartilage. Arthritis Rheum. 60, 2985–2996 (2009).

    Article  CAS  PubMed  Google Scholar 

  85. Porée, B. et al. Interleukin-6 (IL-6) and/or soluble IL-6 receptor down-regulation of human type II collagen gene expression in articular chondrocytes requires a decrease of Sp1.Sp3 ratio and of the binding activity of both factors to the COL2A1 promoter. J. Biol. Chem. 283, 4850–4865 (2008).

    Article  CAS  PubMed  Google Scholar 

  86. Alonzi, T. et al. Interleukin 6 is required for the development of collagen-induced arthritis. J. Exp. Med. 187, 461–468 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. van de Loo, F. A., Kuiper, S., van Enckevort, F. H., Arntz, O. J. & van den Berg, W. B. Interleukin-6 reduces cartilage destruction during experimental arthritis. A study in interleukin-6-deficient mice. Am. J. Pathol. 151, 177–191 (1997).

    CAS  PubMed  PubMed Central  Google Scholar 

  88. de Hooge, A. S. et al. Male IL-6 gene knock out mice developed more advanced osteoarthritis upon aging. Osteoarthritis Cartilage 13, 66–73 (2005).

    Article  PubMed  Google Scholar 

  89. Tilg, H., Dinarello, C. A. & Mier, J. W. IL-6 and APPs: anti-inflammatory and immunosuppressive mediators. Immunol. Today 18, 428–432 (1997).

    Article  CAS  PubMed  Google Scholar 

  90. Kwan Tat, S., Padrines, M., Théoleyre, S., Heymann, D. & Fortun, Y. IL-6, RANKL, TNFα/IL-1: interrelations in bone resorption pathophysiology. Cytokine Growth Factor Rev. 15, 49–60 (2004).

    Article  CAS  PubMed  Google Scholar 

  91. Liu, X. H., Kirschenbaum, A., Yao, S. & Levine, A. C. The role of the interleukin-6/gp130 signaling pathway in bone metabolism. Vitam. Horm. 74, 341–355 (2006).

    Article  CAS  PubMed  Google Scholar 

  92. Massicotte, F. et al. Can altered production of interleukin-1β, interleukin-6, transforming growth factor β and prostaglandin E(2) by isolated human subchondral osteoblasts identify two subgroups of osteoarthritic patients. Osteoarthritis Cartilage 10, 491–500 (2002).

    Article  CAS  PubMed  Google Scholar 

  93. Kwan Tat, S. et al. The differential expression of osteoprotegerin (OPG) and receptor activator of nuclear factor κB ligand (RANKL) in human osteoarthritic subchondral bone osteoblasts is an indicator of the metabolic state of these disease cells. Clin. Exp. Rheumatol. 26, 295–304 (2008).

    CAS  PubMed  Google Scholar 

  94. Chenoufi, H. L. et al. Increased mRNA expression and protein secretion of interleukin-6 in primary human osteoblasts differentiated in vitro from rheumatoid and osteoarthritic bone. J. Cell. Biochem. 81, 666–678 (2001).

    Article  CAS  PubMed  Google Scholar 

  95. Jochems, C., Lagerquist, M., Håkansson, C., Ohlsson, C. & Carlsten, H. Long-term anti-arthritic and anti-osteoporotic effects of raloxifene in established experimental postmenopausal polyarthritis. Clin. Exp. Immunol. 152, 593–597 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Genovese, M. C. et al. Interleukin-6 receptor inhibition with tocilizumab reduces disease activity in rheumatoid arthritis with inadequate response to disease-modifying antirheumatic drugs: the tocilizumab in combination with traditional disease-modifying antirheumatic drug therapy study. Arthritis Rheum. 58, 2968–2980 (2008).

    Article  CAS  PubMed  Google Scholar 

  97. Smolen, J. S. et al. Effect of interleukin-6 receptor inhibition with tocilizumab in patients with rheumatoid arthritis (OPTION study): a double-blind, placebo-controlled, randomised trial. Lancet 371, 987–997 (2008).

    Article  CAS  PubMed  Google Scholar 

  98. Garnero, P., Thompson, E., Woodworth, T. & Smolen, J. S. Rapid and sustained improvement in bone and cartilage turnover markers with the anti-interleukin-6 receptor inhibitor tocilizumab plus methotrexate in rheumatoid arthritis patients with an inadequate response to methotrexate: results from a substudy of the multicenter double-blind, placebo-controlled trial of tocilizumab in inadequate responders to methotrexate alone. Arthritis Rheum. 62, 33–43 (2010).

    Article  CAS  PubMed  Google Scholar 

  99. Doschak, M. R., Wohl, G. R., Hanley, D. A., Bray, R. C. & Zernicke, R. F. Antiresorptive therapy conserves some periarticular bone and ligament mechanical properties after anterior cruciate ligament disruption in the rabbit knee. J. Orthop. Res. 22, 942–948 (2004).

    Article  CAS  PubMed  Google Scholar 

  100. Jones, M. D. et al. In vivo microfocal computed tomography and micro-magnetic resonance imaging evaluation of antiresorptive and antiinflammatory drugs as preventive treatments of osteoarthritis in the rat. Arthritis Rheum. 62, 2726–2735 (2010).

    Article  PubMed  Google Scholar 

  101. Bingham, C. O. 3rd et al. Risedronate decreases biochemical markers of cartilage degradation but does not decrease symptoms or slow radiographic progression in patients with medial compartment osteoarthritis of the knee: results of the two-year multinational knee osteoarthritis structural arthritis study. Arthritis Rheum. 54, 3494–3507 (2006).

    Article  CAS  PubMed  Google Scholar 

  102. Garnero, P. et al. Relationships between biochemical markers of bone and cartilage degradation with radiological progression in patients with knee osteoarthritis receiving risedronate: the Knee Osteoarthritis Structural Arthritis randomized clinical trial. Osteoarthritis Cartilage 16, 660–666 (2008).

    Article  CAS  PubMed  Google Scholar 

  103. Raynauld, J. P. et al. Protective effects of licofelone, a 5-lipoxygenase and cyclo-oxygenase inhibitor, versus naproxen on cartilage loss in knee osteoarthritis: a first multicentre clinical trial using quantitative MRI. Ann. Rheum. Dis. 68, 938–947 (2009).

    Article  CAS  PubMed  Google Scholar 

  104. Scanzello, C. R. et al. Local cytokine profiles in knee osteoarthritis: elevated synovial fluid interleukin-15 differentiates early from end-stage disease. Osteoarthritis Cartilage 17, 1040–1048 (2009).

    Article  CAS  PubMed  Google Scholar 

  105. Martel-Pelletier, J., Mineau, F., Jovanovic, D., Di Battista, J. A. & Pelletier, J. P. Mitogen-activated protein kinase and nuclear factor κB together regulate interleukin-17-induced nitric oxide production in human osteoarthritic chondrocytes: possible role of transactivating factor mitogen-activated protein kinase-activated proten kinase (MAPKAPK). Arthritis Rheum. 42, 2399–2409 (1999).

    Article  CAS  PubMed  Google Scholar 

  106. Lubberts, E., Joosten, L. A., van de Loo, F. A., van den Gersselaar, L. A. & van den Berg, W. B. Reduction of interleukin-17-induced inhibition of chondrocyte proteoglycan synthesis in intact murine articular cartilage by interleukin-4. Arthritis Rheum. 43, 1300–1306 (2000).

    Article  CAS  PubMed  Google Scholar 

  107. Olee, T., Hashimoto, S., Quach, J. & Lotz, M. IL-18 is produced by articular chondrocytes and induces proinflammatory and catabolic responses. J. Immunol. 162, 1096–1100 (1999).

    CAS  PubMed  Google Scholar 

  108. Alaaeddine, N. et al. Differential effects of IL-8, LIF (pro-inflammatory) and IL-11 (anti-inflammatory) on TNF-α-induced PGE2 release and on signalling pathways in human OA synovial fibroblasts. Cytokine 11, 1020–1030 (1999).

    Article  CAS  PubMed  Google Scholar 

  109. Ishii, H. et al. Characterization of infiltrating T cells and TH1/TH2-type cytokines in the synovium of patients with osteoarthritis. Osteoarthritis Cartilage 10, 277–281 (2002).

    Article  CAS  PubMed  Google Scholar 

  110. Alsalameh, S. et al. Cellular immune response toward human articular chondrocytes. T cell reactivities against chondrocyte and fibroblast membranes in destructive joint diseases. Arthritis Rheum. 33, 1477–1486 (1990).

    Article  CAS  PubMed  Google Scholar 

  111. Pacquelet, S. et al. Interleukin 17, a nitric oxide-producing cytokine with a peroxynitrite-independent inhibitory effect on proteoglycan synthesis. J. Rheumatol. 29, 2602–2610 (2002).

    CAS  PubMed  Google Scholar 

  112. Benderdour, M. et al. Interleukin 17 (IL-17) induces collagenase-3 production in human osteoarthritic chondrocytes via AP-1 dependent activation: differential activation of AP-1 members by IL-17 and IL-1β. J. Rheumatol. 29, 1262–1272 (2002).

    CAS  PubMed  Google Scholar 

  113. Honorati, M. C., Bovara, M., Cattini, L., Piacentini, A. & Facchini, A. Contribution of interleukin 17 to human cartilage degradation and synovial inflammation in osteoarthritis. Osteoarthritis Cartilage 10, 799–807 (2002).

    Article  CAS  PubMed  Google Scholar 

  114. Kauffmann, C. et al. Computer-aided method for quantification of cartilage thickness and volume changes using MRI: validation study using a synthetic model. IEEE Trans. Biomed. Eng. 50, 978–988 (2003).

    Article  PubMed  Google Scholar 

  115. Raynauld, J. P. et al. Reliability of a quantification imaging system using magnetic resonance images to measure cartilage thickness and volume in human normal and osteoarthritic knees. Osteoarthritis Cartilage 11, 351–360 (2003).

    Article  PubMed  Google Scholar 

  116. Berthiaume, M. J. et al. Meniscal tear and extrusion are strongly associated with progression of symptomatic knee osteoarthritis as assessed by quantitative magnetic resonance imaging. Ann. Rheum. Dis. 64, 556–563 (2005).

    Article  PubMed  Google Scholar 

  117. Raynauld, J. P. et al. Correlation between bone lesion changes and cartilage volume loss in patients with osteoarthritis of the knee as assessed by quantitative magnetic resonance imaging over a 24-month period. Ann. Rheum. Dis. 67, 683–688 (2008).

    Article  PubMed  Google Scholar 

  118. Pelletier, J. P. et al. A new non-invasive method to assess synovitis severity in relation to symptoms and cartilage volume loss in knee osteoarthritis patients using MRI. Osteoarthritis Cartilage 16 (Suppl. 3), S8–S13 (2008).

    Article  PubMed  Google Scholar 

  119. Li, W. et al. Human hip joint cartilage: MRI quantitative thickness and volume measurements discriminating acetabulum and femoral head. IEEE Trans. Biomed. Eng. 55, 2731–2740 (2008).

    Article  PubMed  Google Scholar 

  120. Li, W. et al. Fully automated system for the quantification of human osteoarthritic knee joint effusion volume using magnetic resonance imaging. Arthritis Res. Ther. 12, R173 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors would like to thank Francois Mineau for his work on the figure for this review article.

Author information

Authors and Affiliations

Authors

Contributions

All authors contributed equally to researching data for the article, providing a substantial contribution to discussions of the content, writing the article, and to the review and/or editing of the manuscript before submission.

Corresponding author

Correspondence to Johanne Martel-Pelletier.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Kapoor, M., Martel-Pelletier, J., Lajeunesse, D. et al. Role of proinflammatory cytokines in the pathophysiology of osteoarthritis. Nat Rev Rheumatol 7, 33–42 (2011). https://doi.org/10.1038/nrrheum.2010.196

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrrheum.2010.196

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing