Skip to main content
Log in

Mechanisms of Chronic Pain in Osteoarthritis

  • RHEUMATIC MANIFESTATIONS OF OTHER DISEASES (R STAUD, SECTION EDITOR)
  • Published:
Current Rheumatology Reports Aims and scope Submit manuscript

Abstract

Pain is a major clinical problem of osteoarthritis (OA). Recently, OA has been thought to be a disease of the whole joint with both destruction of cartilage and inflammatory components such as synovitis and bone marrow lesions. Clinical studies have documented a significant inflammatory soft tissue contribution to the severity and frequency of OA pain. Both clinical and experimental studies have provided evidence for the sensitization of pain pathways during OA, involving pronounced changes in joint nociceptors and changes of the nociceptive processing in the spinal cord, brainstem, and thalamocortical system. Additionally, evidence has been provided for neuropathic pain components in OA models. Concerning molecular mechanisms of OA pain and potential options for pain therapy, studies on nerve growth factor, cytokines, sodium channel blockers, hyaluronic acid preparations, and others are addressed in this review.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1

Similar content being viewed by others

References

Papers of particular interest, published recently, have been highlighted as: • Of importance •• Of major importance

  1. Breivik H, Collett B, Ventafridda V, et al. Survey of chronic pain in Europe: prevalence, impact on daily life, and treatment. Eur J Pain. 2006;10:287–333.

    Article  PubMed  Google Scholar 

  2. Felson DT. Developments in the clinical understanding of osteoarthritis. Arthritis Res Ther. 2009;11:203.

    Article  PubMed  Google Scholar 

  3. Konttinen YT, Sillat T, Barreto G, et al. Osteoarthritis as an autoinflammatory disease caused by chondrocyte-mediated inflammatory responses. Arthritis Rheum. 2012;64:613–6.

    Article  PubMed  CAS  Google Scholar 

  4. Goldring MB, Otero M. Inflammation in osteoarthritis. Curr Opin Rheumatol. 2011;23:471–8.

    Article  PubMed  CAS  Google Scholar 

  5. Bondeson J, Blom AB, Wainwright S. The role of synovial macrophages and macrophage-produced mediators in driving inflammatory and destructive responses in osteoarthritis. Arthritis Rheum. 2010;62:647–57.

    Article  PubMed  CAS  Google Scholar 

  6. • Zhang Y, Nevitt M, Niu J, et al. Fluctuation of knee pain and changes in bone marrow lesions, effusions, and synovitis on magnetic resonance imaging. Arthritis Rheum. 2011;63:691–9. This paper reports important data on the sources of pain in patients with knee OA.

    Article  PubMed  Google Scholar 

  7. Ordeberg G. Evidence of sensitization to pain in human osteoarthritis. In: Felson DT, Schaible H-G, editors. Pain in osteoarthritis. Hoboken: Wiley Blackwell; 2009. p. 199–209.

    Google Scholar 

  8. Hochman JR, Gagliese L, Davis AM, Hawker GA. Neuropathic pain symptoms in a community knee OA cohort. Osteoarthritis Cartilage. 2011;19:647–54.

    Article  PubMed  CAS  Google Scholar 

  9. Eckstein F, Cotofana S, Wirth W, et al. Greater rates of cartilage loss in painful knees than in pain-free knees after adjustment for radiographic disease stage. Arthritis Rheum. 2011;63:2257–67.

    Article  PubMed  Google Scholar 

  10. Wittoek R, Cruyssen BV, Verbruggen G. Predictors of functional impairment and pain in erosive osteoarthritis of the interphalangeal joints. Arthritis Rheum. 2012;64:1430–6.

    Article  PubMed  Google Scholar 

  11. Schaible H-G. Joint pain – Basic mechanisms. In: McMahon SB, Koltzenburg M, editors. Wall and Melzack´s textbook of pain. 6th ed. Oxford: Elsevier; 2012. in press.

    Google Scholar 

  12. Im H-J, Kim J-S, Li X, et al. Alteration of sensory neurons and spinal response to an experimental osteoarthritis pain model. Arthritis Rheum. 2010;62:2995–3005.

    Article  PubMed  CAS  Google Scholar 

  13. Ivanavicius SP, Ball AD, Heapy CG, et al. Structural pathology in a rodent model of osteoarthritis is associated with neuropathic pain: increased expression of ATF-3 and pharmacological characterisation. Pain. 2007;128:272–82.

    Article  PubMed  CAS  Google Scholar 

  14. Orita S, Ishikawa T, Miyagi M, et al. Pain-related sensory innervation in monoiodoacetate-induced osteoarthritis in rat knees that gradually develops neuronal injury in addition to inflammatory pain. BMC Musculoskelet Disord. 2011;12:134.

    Article  PubMed  Google Scholar 

  15. Liu P, Okun A, Guo R-C, et al. Ongoing pain in the MIA model of osteoarthritis. Neurosci Lett. 2011;493:72–5.

    Article  PubMed  CAS  Google Scholar 

  16. Ferland CE, Levrty S, Beaudry F, Vachon P. Gait analysis and pain response of two rodent models of osteoarthritis. Pharmacol Biochem Behav. 2011;97:603–10.

    Article  PubMed  CAS  Google Scholar 

  17. Malfait AM, Ritchie J, Gil AS, et al. ADAMTS-5 deficient mice do not develop mechanical allodynia associated with osteoarthritis following medial meniscal destabilization. Osteoarthritis Cartilage. 2010;18:572–80.

    Article  PubMed  CAS  Google Scholar 

  18. Schaible H-G, Richter F, Ebersberger A, et al. Joint pain. Exp Brain Res. 2009;196:153–62.

    Article  PubMed  Google Scholar 

  19. Schuelert N, McDougall JJ. Grading of monosodium iodoacetate-induced osteoarthritis reveals a concentration-dependent sensitization of nociception in the knee joint of the rat. Neurosci Lett. 2009;465:184–8.

    Article  PubMed  CAS  Google Scholar 

  20. McDougall JJ, Andruski B, Schuelert N, et al. Unravelling the relationship between age, nociception and joint destruction in naturally occurring osteoarthritis of Dunkin Hartley guinea pigs. Pain. 2009;141:222–32.

    Article  PubMed  Google Scholar 

  21. Schaible H-G, Ebersberger A, Natura G. Update on peripheral mechanisms of pain: beyond prostaglandins and cytokines. Arthritis Res Ther. 2011;13:210.

    Article  PubMed  Google Scholar 

  22. •• Lane NE, Schnitzer TJ, Birbara CA, et al. Tanezumab for the treatment of pain from osteoarthritis of the knee. N Engl J Med. 2010;363:1521–31. This study shows for the first time the use of a monoclonal antibody to NGF in pain treatment in patients with moderate to severe OA pain.

    Article  PubMed  CAS  Google Scholar 

  23. Nagashima H, Suzuki M, Araki S, et al. Preliminary assessment of the safety and efficacy of tenazumab in Japanese patients with moderate to severe osteoarthritis of the knee: a randomized, double-blind, dose-escalation, placebo-controlled study. Osteoarthritis Cartilage. 2011;19:1405–12.

    Article  PubMed  CAS  Google Scholar 

  24. Schnitzer TJ, Lane NE, Birbara C, et al. Long-term open-label study of tanezumab for moderate to severe osteoarthritis. Osteoarthritis Cartilage. 2011;19:639–46.

    Article  PubMed  CAS  Google Scholar 

  25. Bennett D. NGF, sensitization of nociceptors. In: Schmidt RF, Willis WD, editors. Encyclopedia of pain, volume 2. Berlin, Heidelberg New York, Tokyo: Springer; 2007. p. 1338–42.

    Chapter  Google Scholar 

  26. McNamee KE, Burleigh A, Gompels LL, et al. Treatment of murine osteoarthritis with TrkAd5 reveals a pivotal role for nerve growth factor in non-inflammatory joint pain. Pain. 2010;149:386–92.

    Article  PubMed  CAS  Google Scholar 

  27. Richter F, Natura G, Loeser S, et al. Tumor necrosis factor-α (TNF-α) causes persistent sensitization of joint nociceptors for mechanical stimuli. Arthritis Rheum. 2010;62:3806–14.

    Article  PubMed  CAS  Google Scholar 

  28. Schaible H-G, Segond von Banchet G, Boettger MK, et al. The role of proinflammatory cytokines in the generation and maintenance of joint pain. Ann N Y Acad Sci. 2010;1193:60–9.

    Article  PubMed  CAS  Google Scholar 

  29. Boettger MK, Leuchtweis J, Kümmel D, et al. Differential effects of locally and systemically administered soluble glycoprotein 130 on pain and inflammation in experimental arthritis. Arthritis Res Ther. 2010;12:R140.

    Article  PubMed  Google Scholar 

  30. Attur M, Belitskaya-Lévy I, Oh C, et al. Increased interleukin-1ß gene expression in peripheral blood leukocytes is associated with increased pain and predicts risk for progression of symptomatic knee osteoarthritis. Arthritis Rheum. 2011;63:1908–17.

    Article  PubMed  CAS  Google Scholar 

  31. Schuelert N, McDougall JJ. Involvement of Nav1.8 sodium channels in the transduction of mechanical pain in a rodent model of osteoarthritis. Arthritis Res Ther. 2012;14:R5.

    Article  PubMed  CAS  Google Scholar 

  32. Schuelert N, Zhang C, Mogg AJ, et al. Paradoxical effects of the cannabinoid CB(2) receptor agonist GW405833 on rat osteoarthritic knee joint pain. Osteoarthritis Cartilage. 2010;18:1536–43.

    Article  PubMed  CAS  Google Scholar 

  33. Zhang W, Robertson J, Jones AC, Dieppe PA, Doherty M. The placebo effect and its determinants in osteoarthritis: meta-analysis of randomised controlled trials. Ann Rheum Dis. 2008;67:1716–23.

    Article  PubMed  CAS  Google Scholar 

  34. Boettger MK, Kümmel D, Harrison A, Schaible H-G. Evaluation of long-term antinociceptive properties of of stabilized hyaluronic acid preparation (NASHA) in an animal model of repetitive joint pain. Arthritis Res Ther. 2011;13:R110.

    Article  PubMed  CAS  Google Scholar 

  35. Jorgensen A, Stengaard-Pedersen K, Simonsen O, et al. Intra-articular hyaluronan is without clinical effect in knee osteoarthritis: a multicentre, randomised, placebo-controlled, double blind study of 337 patients followed for 1 year. Ann Rheum Dis. 2010;69:1097–102.

    Article  PubMed  Google Scholar 

  36. Chevalier X, Jerosch J, Goupille P, et al. Single, intraarticular treatment with 6 ml hylan G-F 20 in patients with symptomatic primary osteoarthritis of the knee: a randomised, multicentre, double-blind, placebo controlled trial. Ann Rheum Dis. 2010;69:113–9.

    Article  PubMed  CAS  Google Scholar 

  37. Berenbaum F, Grifka J, Cazzaniga J, et al. A randomised, double-blind, controlled trial comparing two intra-articular hyaluronic acid preparations differing by their molecular weight in symptomatic knee osteoarthritis. Ann Rheum Dis. doi:10.1136/annrheumdis-2011-200972.

  38. Gomis A, Miralles A, Schmidt RF, Belmonte C. Intra-articular injections of hyaluronan solutions of different elastoviscosity reduce nociceptive nerve activity in a model of osteoarthritic knee joint of the guinea pig. Osteoarthritis Cartilage. 2009;17:798–804.

    Article  PubMed  CAS  Google Scholar 

  39. Ashraf S, Mapp PI, Walsh DA. Contributions of angiogenesis to inflammation, joint damage, and pain in a rat model of osteoarthritis. Arthritis Rheum. 2011;63:2700–10.

    Article  PubMed  CAS  Google Scholar 

  40. Arendt-Nielsen L, Nie H, Laursen MB, et al. Sensitization in patients with painful knee osteoarthritis. Pain. 2010;149:573–81.

    Article  PubMed  Google Scholar 

  41. Rahman W, Bauer CS, Bannister K, et al. Descending serotoninergic facilitation and the antinociceptive effects of pregabalin in a rat model of osteoarthitic pain. Mol Pain. 2009;5:45.

    Article  PubMed  Google Scholar 

  42. Sagar DR, Staniaszek LE, Okine BN, et al. Tonic modulation of spinal hyperexcitability by the endocannabinoid receptor system in a rat model of osteoarthritis pain. Arthritis Rheum. 2010;62:3666–76.

    Article  PubMed  CAS  Google Scholar 

  43. Telleria-Diaz A, Schmidt M, Kreusch S, et al. Spinal antinociceptive effects of cyclooxygenase inhibition during inflammation: involvement of prostaglandins and endocannabinoids. Pain. 2010;148:26–35.

    Article  PubMed  CAS  Google Scholar 

  44. Kosek E, Ordeberg G. Lack of pressure pain modulation by heterotopic noxious conditioning stimulation in patients with painful osteoarthritis before, but not following surgical pain relief. Pain. 2000;88:69–78.

    Article  PubMed  CAS  Google Scholar 

  45. Gwilym SE, Keltner JR, Warnaby CE, et al. Psychophysical and functional imaging evidence supporting the presence of central sensitization in a cohort of osteoarthritis patients. Arthritis Rheum. 2009;61:1226–34.

    Article  PubMed  Google Scholar 

  46. Kulkarni B, Bentley DE, Elliott R, et al. Arthritic pain is processed in brain areas concerned with emotions and fear. Arthritis Rheum. 2007;56:1345–54.

    Article  PubMed  CAS  Google Scholar 

  47. Gwilym SE, Filippini N, Douaud G, et al. Thalamic atrophy associated with painful osteoarthritis of the hip is reversible after arthroplasty. Arthritis Rheum. 2010;62:2930–40.

    Article  PubMed  Google Scholar 

  48. Rodriguez-Raecke R, Niemeier A, Ihle K, et al. Brain gray matter decrease in chronic pain is the consequence and not the cause of pain. J Neurosci. 2009;29:13746–50.

    Article  PubMed  CAS  Google Scholar 

  49. Hess A, Axmann R, Rech J, et al. Blockade of TNF-α rapidly inhibits pain responses in the central nervous system. PNAS. 2011;108:3731–36.

    Article  PubMed  CAS  Google Scholar 

Download references

Disclosure

No potential conflicts of interest relevant to this article were reported.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Hans-Georg Schaible.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Schaible, HG. Mechanisms of Chronic Pain in Osteoarthritis. Curr Rheumatol Rep 14, 549–556 (2012). https://doi.org/10.1007/s11926-012-0279-x

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11926-012-0279-x

Keywords

Navigation