MMP-13 in Osteoarthritis Treatment: Comparison
Please note this is a comparison between Version 2 by Conner Chen and Version 1 by Melanie Ecker.

Osteoarthritis (OA) is a common degenerative disease characterized by the destruction of articular cartilage and chronic inflammation of surrounding tissues. Matrix metalloproteinase-13 (MMP-13) is the primary MMP involved in cartilage degradation through its particular ability to cleave type II collagen. Hence, it is an attractive target for the treatment of OA.

  • osteoarthritis
  • MMP-13
  • inhibitor
Please wait, diff process is still running!

References

  1. Castrogiovanni, P.; Musumeci, G. Which is the best physical treatment for osteoarthritis? J. Funct. Morphol. Kinesiol. 2016, 1, 54–68.
  2. Litwic, A.; Edwards, M.H.; Dennison, E.M.; Cooper, C. Epidemiology and burden of osteoarthritis. Br. Med. Bull. 2013, 105, 185–199.
  3. James, S.L.; Abate, D.; Abate, K.H.; Abay, S.M.; Abbafati, C.; Abbasi, N.; Abbastabar, H.; Abd-Allah, F.; Abdela, J.; Abdelalim, A.; et al. Global, regional, and national incidence, prevalence, and years lived with disability for 354 Diseases and Injuries for 195 countries and territories, 1990-2017: A systematic analysis for the Global Burden of Disease Study 2017. Lancet 2018.
  4. Wilder, F.V.; Hall, B.J.; Barrett, J.P.; Lemrow, N.B. History of acute knee injury and osteoarthritis of the knee: A prospective epidemiological assessment. The clearwater osteoarthritis study. Osteoarthr. Cartil. 2002, 10, 611–616.
  5. Niu, J.; Zhang, Y.Q.; Torner, J.; Nevitt, M.; Lewis, C.E.; Aliabadi, P.; Sack, B.; Clancy, M.; Sharma, L.; Felson, D.T. Is obesity a risk factor for progressive radiographic knee osteoarthritis? Arthritis Care Res. 2009, 61, 329–335.
  6. Palazzo, C.; Nguyen, C.; Lefevre-Colau, M.M.; Rannou, F.; Poiraudeau, S. Risk factors and burden of osteoarthritis. Ann. Phys. Rehabil. Med. 2016, 59, 134–138.
  7. Donell, S. Subchondral bone remodelling in osteoarthritis. EFORT Open Rev. 2019, 4, 221–229.
  8. Melrose, J.; Fuller, E.S.; Little, C.B. The biology of meniscal pathology in osteoarthritis and its contribution to joint disease: Beyond simple mechanics. Connect. Tissue Res. 2017, 58, 282–294.
  9. Belluzzi, E.; Stocco, E.; Pozzuoli, A.; Granzotto, M.; Porzionato, A.; Vettor, R.; De Caro, R.; Ruggieri, P.; Ramonda, R.; Rossato, M.; et al. Contribution of Infrapatellar Fat Pad and Synovial Membrane to Knee Osteoarthritis Pain. Biomed Res. Int. 2019, 2019.
  10. Zeng, N.; Yan, Z.P.; Chen, X.Y.; Ni, G.X. Infrapatellar fat pad and knee osteoarthritis. Aging Dis. 2020, 11, 1317–1328.
  11. Malemud, C.J. Biologic basis of osteoarthritis: State of the evidence. Curr. Opin. Rheumatol. 2015, 27, 289–294.
  12. Martel-Pelletier, J.; Barr, A.J.; Cicuttini, F.M.; Conaghan, P.G.; Cooper, C.; Goldring, M.B.; Goldring, S.R.; Jones, G.; Teichtahl, A.J.; Pelletier, J.P. Osteoarthritis. Nat. Rev. Dis. Prim. 2016, 2.
  13. Takaishi, H.; Kimura, T.; Dalal, S.; Okada, Y.; D’Armiento, J. Joint Diseases and Matrix Metalloproteinases: A Role for MMP-13. Curr. Pharm. Biotechnol. 2008, 9, 47–54.
  14. Goldring, M.B. The role of the chondrocyte in osteoarthritis. Arthritis Rheum. 2000, 43, 1916–1926.
  15. Man, G.S.; Mologhianu, G. Osteoarthritis pathogenesis—A complex process that involves the entire joint. J. Med. Life 2014, 7, 37–41.
  16. Zhang, F.J.; Yu, W.B.; Luo, W.; Gao, S.G.; Li, Y.S.; Lei, G.H. Effect of osteopontin on TIMP-1 and TIMP-2 mRNA in chondrocytes of human knee osteoarthritis in vitro. Exp. Ther. Med. 2014, 8, 391–394.
  17. Mitchell, P.G.; Magna, H.A.; Reeves, L.M.; Lopresti-Morrow, L.L.; Yocum, S.A.; Rosner, P.J.; Geoghegan, K.F.; Hambor, J.E. Cloning, expression, and type II collagenolytic activity of matrix metalloproteinase-13 from human osteoarthritic cartilage. J. Clin. Investing. 1996, 97, 761–768.
  18. Mueller, M.B.; Tuan, R.S. Anabolic/Catabolic balance in pathogenesis of osteoarthritis: Identifying molecular targets. PM R 2011, 3, S3–S11.
  19. Di Rosa, M.; Castrogiovanni, P.; Musumeci, G. The synovium theory: Can exercise prevent knee osteoarthritis? The role of “mechanokines”, a possible biological key. J. Funct. Morphol. Kinesiol. 2019, 4, 11.
  20. Castrogiovanni, P.; Di Rosa, M.; Ravalli, S.; Castorina, A.; Guglielmino, C.; Imbesi, R.; Vecchio, M.; Drago, F.; Szychlinska, M.A.; Musumeci, G. Moderate physical activity as a prevention method for knee osteoarthritis and the role of synoviocytes as biological key. Int. J. Mol. Sci. 2019, 20, 511.
  21. Chow, Y.Y.; Chin, K.Y. The Role of Inflammation in the Pathogenesis of Osteoarthritis. Mediators Inflamm. 2020.
  22. Sinusas, K. Osteoarthritis: Diagnosis and treatment. Am. Fam. Physician 2012, 85, 49–56.
  23. Flood, J. The role of acetaminophen in the treatment of osteoarthritis. Am. J. Manag. Care 2010, 16, S48–S54.
  24. Darwiche, H.; Barsoum, W.K.; Klika, A.; Krebs, V.E.; Molloy, R. Retrospective analysis of infection rate after early reoperation in total hip arthroplasty. Proc. Clin. Orthop. Relat. Res. 2010, 468, 2392–2396.
  25. Mahler, E.A.M.; Minten, M.J.M.; Leseman-Hoogenboom, M.M.; Poortmans, P.M.P.; Leer, J.W.H.; Boks, S.S.; Van Den Hoogen, F.H.J.; Den Broeder, A.A.; Van Den Ende, C.H.M. Effectiveness of low-dose radiation therapy on symptoms in patients with knee osteoarthritis: A randomised, double-blinded, sham-controlled trial. Ann. Rheum. Dis. 2019, 78, 83–90.
  26. Stevens, R.M.; Ervin, J.; Nezzer, J.; Nieves, Y.; Guedes, K.; Burges, R.; Hanson, P.D.; Campbell, J.N. Randomized, Double-Blind, Placebo-Controlled Trial of Intraarticular Trans-Capsaicin for Pain Associated with Osteoarthritis of the Knee. Arthritis Rheumatol. 2019, 71, 1524–1533.
  27. Kloppenburg, M.; Peterfy, C.; Haugen, I.K.; Kroon, F.; Chen, S.; Wang, L.; Liu, W.; Levy, G.; Fleischmann, R.M.; Berenbaum, F.; et al. Phase IIa, placebo-controlled, randomised study of lutikizumab, an anti-interleukin-1α and anti-interleukin-1β dual variable domain immunoglobulin, in patients with erosive hand osteoarthritis. Ann. Rheum. Dis. 2019, 78, 413–420.
  28. Jin, Y.; Smith, C.; Monteith, D.; Brown, R.; Camporeale, A.; McNearney, T.A.; Deeg, M.A.; Raddad, E.; Xiao, N.; de la Peña, A.; et al. CGRP blockade by galcanezumab was not associated with reductions in signs and symptoms of knee osteoarthritis in a randomized clinical trial. Osteoarthr. Cartil. 2018, 26, 1609–1618.
  29. Jo, C.H.; Lee, Y.G.; Shin, W.H.; Kim, H.; Chai, J.W.; Jeong, E.C.; Kim, J.E.; Shim, H.; Shin, J.S.; Shin, I.S.; et al. Intra-articular injection of mesenchymal stem cells for the treatment of osteoarthritis of the knee: A proof-of-concept clinical trial. Stem Cells 2014, 32, 1254–1266.
  30. Meheux, C.J.; McCulloch, P.C.; Lintner, D.M.; Varner, K.E.; Harris, J.D. Efficacy of Intra-articular Platelet-Rich Plasma Injections in Knee Osteoarthritis: A Systematic Review. Arthrosc. J. Arthrosc. Relat. Surg. 2016, 32, 495–505.
  31. Sabatini, M.; Lesur, C.; Thomas, M.; Chomel, A.; Anract, P.; De Nanteuil, G.; Pastoureau, P. Effect of inhibition of matrix metalloproteinases on cartilage loss in vitro and in a guinea pig model of osteoarthritis. Arthritis Rheum. 2005, 52, 171–180.
  32. Renkiewicz, R.; Qiu, L.; Lesch, C.; Sun, X.; Devalaraja, R.; Cody, T.; Kaldjian, E.; Welgus, H.; Baragi, V. Broad-spectrum matrix metalloproteinase inhibitor marimastat-induced musculoskeletal side effects in rats. Arthritis Rheum. 2003, 48, 1742–1749.
  33. Xie, X.W.; Wan, R.Z.; Liu, Z.P. Recent Research Advances in Selective Matrix Metalloproteinase-13 Inhibitors as Anti-Osteoarthritis Agents. ChemMedChem. 2017, 12, 1157–1168.
  34. Whittaker, M.; Floyd, C.D.; Brown, P.; Gearing, A.J.H. Design and Therapeutic Application of Matrix Metalloproteinase Inhibitors. Chem. Rev. 1999, 99, 2735–2776.
  35. Jacobsen, J.A.; Major Jourden, J.L.; Miller, M.T.; Cohen, S.M. To bind zinc or not to bind zinc: An examination of innovative approaches to improved metalloproteinase inhibition. Biochim. Biophys. Acta (BBA) Mol. Cell Res. 2010, 1803, 72–94.
  36. Dufour, A.; Sampson, N.S.; Zucker, S.; Cao, J. Role of the hemopexin domain of matrix metalloproteinases in cell migration. J. Cell. Physiol. 2008, 217, 643–651.
  37. Laronha, H.; Caldeira, J. Structure and Function of Human Matrix Metalloproteinases. Cells 2020, 9, 1076.
  38. MacColl, E.; Khalil, R.A. Matrix metalloproteinases as regulators of vein structure and function: Implications in chronic venous disease. J. Pharmacol. Exp. Ther. 2015, 355, 410–428.
  39. Aureli, L.; Gioia, M.; Cerbara, I.; Monaco, S.; Fasciglione, G.; Marini, S.; Ascenzi, P.; Topai, A.; Coletta, M. Structural Bases for Substrate and Inhibitor Recognition by Matrix Metalloproteinases. Curr. Med. Chem. 2008, 15, 2192–2222.
  40. Overall, C.M.; Kleifeld, O. Towards third generation matrix metalloproteinase inhibitors for cancer therapy. Br. J. Cancer 2006, 94, 941–946.
  41. Park, H.I.; Jin, Y.; Hurst, D.R.; Monroe, C.A.; Lee, S.; Schwartz, M.A.; Sang, Q.X.A. The Intermediate S1′ Pocket of the Endometase/Matrilysin-2 Active Site Revealed by Enzyme Inhibition Kinetic Studies, Protein Sequence Analyses, and Homology Modeling. J. Biol. Chem. 2003, 278, 51646–51653.
  42. Lovejoy, B.; Welch, A.R.; Carr, S.; Luong, C.; Broka, C.; Hendricks, R.T.; Campbell, J.A.; Walker, K.A.M.; Martin, R.; Van Wart, H.; et al. Crystal structures of MMP-1 and -13 reveal the structural basis for selectivity of collagenase inhibitors. Nat. Struct. Biol. 1999, 6, 217–221.
  43. Cui, N.; Hu, M.; Khalil, R.A. Biochemical and Biological Attributes of Matrix Metalloproteinases. Prog. Mol. Biol. Transl. Sci. 2017, 147, 1–73.
  44. Van Wart, H.E.; Birkedal-Hansen, H. The cysteine switch: A principle of regulation of metalloproteinase activity with potential applicability to the entire matrix metalloproteinase gene family. Proc. Natl. Acad. Sci. USA. 1990, 87, 5578–5582.
  45. Nagase, H.; Visse, R.; Murphy, G. Structure and function of matrix metalloproteinases and TIMPs. Cardiovasc. Res. 2006, 69, 562–573.
  46. Knäuper, V.; Will, H.; López-Otin, C.; Smith, B.; Atkinson, S.J.; Stanton, H.; Hembry, R.M.; Murphy, G. Cellular mechanisms for human procollagenase-3 (MMP-13) activation. Evidence that MT1-MMP (MMP-14) and gelatinase A (MMP-2) are able to generate active enzyme. J. Biol. Chem. 1996, 271, 17124–17131.
  47. Zijlstrat, A.; Aimes, R.T.; Zhu, D.; Regazzoni, K.; Kupriyanova, T.; Seandel, M.; Deryugina, E.I.; Quigley, J.P. Collagenolysis-dependent angiogenesis mediated by matrix metalloproteinase-13 (collagenase-3). J. Biol. Chem. 2004, 279, 27633–27645.
  48. Rowan, A.D.; Litherland, G.J.; Hui, W.; Milner, J.M. Metalloproteases as potential therapeutic targets in arthritis treatment. Expert Opin. Ther. Targets 2008, 12, 1–18.
  49. Howes, J.M.; Bihan, D.; Slatter, D.A.; Hamaia, S.W.; Packman, L.C.; Knauper, V.; Visse, R.; Farndale, R.W. The recognition of collagen and triple-helical toolkit peptides by MMP-13: Sequence specificity for binding and cleavage. J. Biol. Chem. 2014, 289, 24091–24101.
  50. Little, C.B.; Barai, A.; Burkhardt, D.; Smith, S.M.; Fosang, A.J.; Werb, Z.; Shah, M.; Thompson, E.W. Matrix metalloproteinase 13-deficient mice are resistant to osteoarthritic cartilage erosion but not chondrocyte hypertrophy or osteophyte development. Arthritis Rheum. 2009, 60, 3723–3733.
  51. Neuhold, L.A.; Killar, L.; Zhao, W.; Sung, M.L.A.; Warner, L.; Kulik, J.; Turner, J.; Wu, W.; Billinghurst, C.; Meijers, T.; et al. Postnatal expression in hyaline cartilage of constitutively active human collagenase-3 (MMP-13) induces osteoarthritis in mice. J. Clin. Invest. 2001, 107, 35–44.
  52. Shiomi, T.; Lemaître, V.; D’Armiento, J.; Okada, Y. Matrix metalloproteinases, a disintegrin and metalloproteinases, and a disintegrin and metalloproteinases with thrombospondin motifs in non-neoplastic diseases: Review Article. Pathol. Int. 2010, 60, 477–496.
  53. Li, H.; Wang, D.; Yuan, Y.; Min, J. New insights on the MMP-13 regulatory network in the pathogenesis of early osteoarthritis. Arthritis Res. Ther. 2017, 19, 1–12.
  54. Wang, X.; Khalil, R.A. Matrix Metalloproteinases, Vascular Remodeling, and Vascular Disease. Adv. Pharmacol. 2018, 81, 241–330.
  55. Troeberg, L.; Nagase, H. Analysis of TIMP expression and activity. Methods Mol. Med. 2007, 135, 251–267.
  56. Brew, K.; Dinakarpandian, D.; Nagase, H. Tissue inhibitors of metalloproteinases: Evolution, structure and function. Biochim. Biophys. Acta 2000, 1477, 267–283.
  57. Sahebjam, S.; Khokha, R.; Mort, J.S. Increased collagen and aggrecan degradation with age in the joints of Timp3-/-mice. Arthritis Rheum. 2007, 56, 905–909.
  58. Black, R.A.; Castner, B.; Slack, J.; Tocker, J.; Eisenman, J.; Jacobson, E.; Delaney, J.; Winters, D.; Hecht, R.; Bendele, A. A14 Injected Timp-3 protects cartilage in a rat meniscal tear model. Osteoarthr. Cartil. 2006, 14, S23–S24.
  59. Kevorkian, L.; Young, D.A.; Darrah, C.; Donell, S.T.; Shepstone, L.; Porter, S.; Brockbank, S.M.V.; Edwards, D.R.; Parker, A.E.; Clark, I.M. Expression profiling of metalloproteinases and their inhibitors in cartilage. Arthritis Rheum. 2004, 50, 131–141.
  60. Strickland, D.K.; Ashcom, J.D.; Williams, S.; Burgess, W.H.; Migliorini, M.; Scott Argraves, W. Sequence identity between the α2-macroglobulin receptor and low density lipoprotein receptor-related protein suggests that this molecule is a multifunctional receptor. J. Biol. Chem. 1990, 265, 17401–17404.
  61. Chen, D.; Kim, D.J.; Shen, J.; Zou, Z.; O’Keefe, R.J. Runx2 plays a central role in Osteoarthritis development. J. Orthop. Transl. 2020, 23, 132–139.
  62. Hirata, M.; Kugimiya, F.; Fukai, A.; Saito, T.; Yano, F.; Ikeda, T.; Mabuchi, A.; Sapkota, B.R.; Akune, T.; Nishida, N.; et al. C/EBPβ and RUNX2 cooperate to degrade cartilage with MMP-13 as the target and HIF-2α as the inducer in chondrocytes. Hum. Mol. Genet. 2012, 21, 1111–1123.
  63. Yun, K.; Im, S.H. Transcriptional regulation of MMP13 by Lef1 in chondrocytes. Biochem. Biophys. Res. Commun. 2007, 364, 1009–1014.
  64. Yun, K.; So, J.-S.; Jash, A.; Im, S.-H. Lymphoid Enhancer Binding Factor 1 Regulates Transcription through Gene Looping. J. Immunol. 2009, 183, 5129–5137.
  65. Grall, F.; Gu, X.; Tan, L.; Cho, J.Y.; Inan, M.S.; Pettit, A.R.; Thamrongsak, U.; Choy, B.K.; Manning, C.; Akbarali, Y.; et al. Responses to the proinflammatory cytokines interleukin-1 and tumor necrosis factor α in cells derived from rheumatoid synovium and other joint tissues involve nuclear factor κB-mediated induction of the Ets transcription factor ESE-1. Arthritis Rheum. 2003, 48, 1249–1260.
  66. Otero, M.; Plumb, D.A.; Tsuchimochi, K.; Dragomir, C.L.; Hashimoto, K.; Peng, H.; Olivotto, E.; Bevilacqua, M.; Tan, L.; Yang, Z.; et al. E74-like Factor 3 (ELF3) impacts on Matrix Metalloproteinase 13 (MMP13) transcriptional control in articular chondrocytes under proinflammatory stress. J. Biol. Chem. 2012, 287, 3559–3572.
  67. Sugita, S.; Hosaka, Y.; Okada, K.; Mori, D.; Yano, F.; Kobayashi, H.; Taniguchi, Y.; Mori, Y.; Okuma, T.; Chang, S.H.; et al. Transcription factor Hes1 modulates osteoarthritis development in cooperation with calcium/calmodulin-dependent protein kinase 2. Proc. Natl. Acad. Sci. USA. 2015, 112, 3080–3085.
  68. Pombo-Suarez, M.; Castaño-Oreja, M.T.; Calaza, M.; Gomez-Reino, J.; Gonzalez, A. Differential upregulation of the three transforming growth factor beta isoforms in human osteoarthritic cartilage. Ann. Rheum. Dis. 2009, 68, 568–571.
  69. Aref-Eshghi, E.; Liu, M.; Harper, P.E.; Doré, J.; Martin, G.; Furey, A.; Green, R.; Rahman, P.; Zhai, G. Overexpression of MMP13 in human osteoarthritic cartilage is associated with the SMAD-independent TGF-β signalling pathway. Arthritis Res. Ther. 2015, 17.
  70. Massicotte, F.; Aubry, I.; Martel-Pelletier, J.; Pelletier, J.P.; Fernandes, J.; Lajeunesse, D. Abnormal insulin-like growth factor 1 signaling in human osteoarthritic subchondral bone osteoblasts. Arthritis Res. Ther. 2006, 8.
  71. Zhang, M.; Zhou, Q.; Liang, Q.Q.; Li, C.G.; Holz, J.D.; Tang, D.; Sheu, T.J.; Li, T.F.; Shi, Q.; Wang, Y.J. IGF-1 regulation of type II collagen and MMP-13 expression in rat endplate chondrocytes via distinct signaling pathways. Osteoarthr. Cartil. 2009, 17, 100–106.
  72. Dvir-Ginzberg, M.; Gagarina, V.; Lee, E.J.; Booth, R.; Gabay, O.; Hall, D.J. Tumor necrosis factor α-mediated cleavage and inactivation of sirT1 in human osteoarthritic chondrocytes. Arthritis Rheum. 2011, 63, 2363–2373.
  73. Elayyan, J.; Lee, E.J.; Gabay, O.; Smith, C.A.; Qiq, O.; Reich, E.; Mobasheri, A.; Henrotin, Y.; Kimber, S.J.; Dvir-Ginzberg, M. LEF1-mediated MMP13 gene expression is repressed by SIRT1 in human chondrocytes. FASEB J. 2017, 31, 3116–3125.
  74. Liu, S.; Yang, H.; Hu, B.; Zhang, M. Sirt1 regulates apoptosis and extracellular matrix degradation in resveratrol-treated osteoarthritis chondrocytes via the wnt/β-catenin signaling pathways. Exp. Ther. Med. 2017, 14, 5057–5062.
  75. Polur, I.; Lee, P.L.; Servais, J.M.; Xu, L.; Li, Y. Role of HTRA1, a serine protease, in the progression of articular cartilage degeneration. Histol. Histopathol. 2010, 25, 599–608.
  76. Xu, L.; Servais, J.; Polur, I.; Kim, D.; Lee, P.L.; Chung, K.; Li, Y. Attenuation of osteoarthritis progression by reduction of discoidin domain receptor 2 in mice. Arthritis Rheum. 2010, 62, 2736–2744.
  77. Yamamoto, K.; Okano, H.; Miyagawa, W.; Visse, R.; Shitomi, Y.; Santamaria, S.; Dudhia, J.; Troeberg, L.; Strickland, D.K.; Hirohata, S.; et al. MMP-13 is constitutively produced in human chondrocytes and co-endocytosed with ADAMTS-5 and TIMP-3 by the endocytic receptor LRP1. Matrix Biol. 2016, 56, 57–73.
  78. Park, D.R.; Kim, J.; Kim, G.M.; Lee, H.; Kim, M.; Hwang, D.; Lee, H.; Kim, H.S.; Kim, W.; Park, M.C.; et al. Osteoclast-associated receptor blockade prevents articular cartilage destruction via chondrocyte apoptosis regulation. Nat. Commun. 2020, 11.
  79. Loeser, R.F. Integrins and chondrocyte-matrix interactions in articular cartilage. Matrix Biol. 2014, 39, 11–16.
  80. Forsyth, C.B.; Pulai, J.; Loeser, R.F. Fibronectin fragments and blocking antibodies to α2β1 and α5β1 integrins stimulate mitogen-activated protein kinase signaling and increase collagenase 3 (matrix metalloproteinase 13) production by human articular chondrocytes. Arthritis Rheum. 2002, 46, 2368–2376.
  81. Hui, W.; Young, D.A.; Rowan, A.D.; Xu, X.; Cawston, T.E.; Proctor, C.J. Oxidative changes and signalling pathways are pivotal in initiating age-related changes in articular cartilage. Ann. Rheum. Dis. 2016, 75, 449–458.
  82. Blaney Davidson, E.N.; Remst, D.F.G.; Vitters, E.L.; van Beuningen, H.M.; Blom, A.B.; Goumans, M.-J.; van den Berg, W.B.; van der Kraan, P.M. Increase in ALK1/ALK5 Ratio as a Cause for Elevated MMP-13 Expression in Osteoarthritis in Humans and Mice. J. Immunol. 2009, 182, 7937–7945.
  83. Wang, W.; Wang, L.; Xu, Z.; Yin, Y.; Su, J.; Niu, X.; Cao, X. Effects of estradiol on reduction of osteoarthritis in rabbits through effect on matrix metalloproteinase proteins. Iran. J. Basic Med. Sci. 2016, 19, 310–315.
  84. Iliopoulos, D.; Malizos, K.N.; Tsezou, A. Epigenetic regulation of leptin affects MMP-13 expression in osteoarthritic chondrocytes: Possible molecular target for osteoarthritis therapeutic intervention. Ann. Rheum. Dis. 2007, 66, 1616–1621.
  85. Francin, P.J.; Abot, A.; Guillaume, C.; Moulin, D.; Bianchi, A.; Gegout-Pottie, P.; Jouzeau, J.Y.; Mainard, D.; Presle, N. Association between adiponectin and cartilage degradation in human osteoarthritis. Osteoarthr. Cartil. 2014, 22, 519–526.
  86. Yammani, R.R.; Loeser, R.F. Brief report: Stress-inducible nuclear protein 1 regulates matrix metalloproteinase 13 expression in human articular chondrocytes. Arthritis Rheumatol. 2014, 66, 1266–1271.
  87. Liang, Y.; Duan, L.; Xiong, J.; Zhu, W.; Liu, Q.; Wang, D.; Liu, W.; Li, Z.; Wang, D. E2 regulates MMP-13 via targeting miR-140 in IL-1β-induced extracellular matrix degradation in human chondrocytes. Arthritis Res. Ther. 2016, 18.
  88. Romanoski, C.E.; Glass, C.K.; Stunnenberg, H.G.; Wilson, L.; Almouzni, G. Epigenomics: Roadmap for regulation. Nature 2015, 518, 314–316.
  89. Bird, A. Perceptions of epigenetics. Nature 2007, 447, 396–398.
  90. Lev Maor, G.; Yearim, A.; Ast, G. The alternative role of DNA methylation in splicing regulation. Trends Genet. 2015, 31, 274–280.
  91. Reynard, L.N.; Loughlin, J. Genetics and epigenetics of osteoarthritis. Maturitas 2012, 71, 200–204.
  92. Hashimoto, K.; Otero, M.; Imagawa, K.; De Andrés, M.C.; Coico, J.M.; Roach, H.I.; Oreffo, R.O.C.; Marcu, K.B.; Goldring, M.B. Regulated transcription of human matrix metalloproteinase 13 (MMP13) and interleukin-1β (IL1B) genes in chondrocytes depends on methylation of specific proximal promoter CpG sites. J. Biol. Chem. 2013, 288, 10061–10072.
  93. Roach, H.I.; Yamada, N.; Cheung, K.S.C.; Tilley, S.; Clarke, N.M.P.; Oreffo, R.O.C.; Kokubun, S.; Bronner, F. Association between the abnormal expression of matrix-degrading enzymes by human osteoarthritic chondrocytes and demethylation of specific CpG sites in the promoter regions. Arthritis Rheum. 2005, 52, 3110–3124.
  94. Cheung, K.S.C.; Hashimoto, K.; Yamada, N.; Roach, H.I. Expression of ADAMTS-4 by chondrocytes in the surface zone of human osteoarthritic cartilage is regulated by epigenetic DNA de-methylation. Rheumatol. Int. 2009, 29, 525–534.
  95. Takahashi, A.; de Andrés, M.C.; Hashimoto, K.; Itoi, E.; Otero, M.; Goldring, M.B.; Oreffo, R.O.C. DNA methylation of the RUNX2 P1 promoter mediates MMP13 transcription in chondrocytes. Sci. Rep. 2017, 7.
  96. Kouzarides, T. Chromatin Modifications and Their Function. Cell 2007, 128, 693–705.
  97. Khan, N.M.; Haqqi, T.M. Epigenetics in osteoarthritis: Potential of HDAC inhibitors as therapeutics. Pharmacol. Res. 2018, 128, 73–79.
  98. Clayton, A.L.; Hazzalin, C.A.; Mahadevan, L.C. Enhanced Histone Acetylation and Transcription: A Dynamic Perspective. Mol. Cell 2006, 23, 289–296.
  99. Hong, S.; Derfoul, A.; Pereira-Mouries, L.; Hall, D.J. A novel domain in histone deacetylase 1 and 2 mediates repression of cartilage-specific genes in human chondrocytes. FASEB J. 2009, 23, 3539–3552.
  100. Higashiyama, R.; Miyaki, S.; Yamashita, S.; Yoshitaka, T.; Lindman, G.; Ito, Y.; Sasho, T.; Takahashi, K.; Lotz, M.; Asahara, H. Correlation between MMP-13 and HDAC7 expression in human knee osteoarthritis. Mod. Rheumatol. 2010, 20, 11–17.
  101. Carpio, L.R.; Bradley, E.W.; Westendorf, J.J. Histone deacetylase 3 suppresses Erk phosphorylation and matrix metalloproteinase (Mmp)-13 activity in chondrocytes. Connect. Tissue Res. 2017, 58, 27–36.
  102. Ma, L.; Bajic, V.B.; Zhang, Z. On the classification of long non-coding RNAs. RNA Biol. 2013, 10, 924–933.
  103. Mattick, J.S.; Makunin, I.V. Non-coding RNA. Hum. Mol. Genet. 2006, 15, R17–R29.
  104. Barter, M.J.; Young, D.A. Epigenetic mechanisms and non-coding rnas in osteoarthritis. Curr. Rheumatol. Rep. 2013, 15.
  105. Sondag, G.R.; Haqqi, T.M. The Role of MicroRNAs and Their Targets in Osteoarthritis. Curr. Rheumatol. Rep. 2016, 18, 56.
  106. Bartel, D.P. MicroRNAs: Genomics, Biogenesis, Mechanism, and Function. Cell 2004, 116, 281–297.
  107. Eulalio, A.; Huntzinger, E.; Nishihara, T.; Rehwinkel, J.; Fauser, M.; Izaurralde, E. Deadenylation is a widespread effect of miRNA regulation. RNA 2009, 15, 21–32.
  108. Asahara, H. Current Status and Strategy of microRNA Research for Cartilage Development and Osteoarthritis Pathogenesis. J. Bone Metab. 2016, 23, 121.
  109. Zhang, H.; Song, B.; Pan, Z. Downregulation of microRNA-9 increases Matrix metalloproteinase-13 expression levels and facilitates osteoarthritis onset. Mol. Med. Rep. 2018, 17, 3708–3714.
  110. Gu, R.; Liu, N.; Luo, S.; Huang, W.; Zha, Z.; Yang, J. MicroRNA-9 regulates the development of knee osteoarthritis through the NF-kappaB1 pathway in chondrocytes. Medicine 2016, 95, e4315.
  111. Song, J.; Kim, D.; Chun, C.H.; Jin, E.J. MicroRNA-9 regulates survival of chondroblasts and cartilage integrity by targeting protogenin. Cell Commun. Signal. 2013, 11.
  112. Yamasaki, K.; Nakasa, T.; Miyaki, S.; Ishikawa, M.; Deie, M.; Adachi, N.; Yasunaga, Y.; Asahara, H.; Ochi, M. Expression of microRNA-146a in osteoarthritis cartilage. Arthritis Rheum. 2009, 60, 1035–1041.
  113. Meng, F.; Zhang, Z.; Chen, W.; Huang, G.; He, A.; Hou, C.; Long, Y.; Yang, Z.; Zhang, Z.; Liao, W. MicroRNA-320 regulates matrix metalloproteinase-13 expression in chondrogenesis and interleukin-1β-induced chondrocyte responses. Osteoarthr. Cartil. 2016, 24, 932–941.
  114. Park, S.J.; Cheon, E.J.; Lee, M.H.; Kim, H.A. MicroRNA-127-5p regulates matrix metalloproteinase 13 expression and interleukin-1β-induced catabolic effects in human chondrocytes. Arthritis Rheum. 2013, 65, 3141–3152.
  115. Akhtar, N.; Rasheed, Z.; Ramamurthy, S.; Anbazhagan, A.N.; Voss, F.R.; Haqqi, T.M. MicroRNA-27b regulates the expression of matrix metalloproteinase 13 in human osteoarthritis chondrocytes. Arthritis Rheum. 2010, 62, 1361–1371.
  116. Wang, G.; Zhang, Y.; Zhao, X.; Meng, C.; Ma, L.; Kong, Y. MicroRNA-411 inhibited matrix metalloproteinase 13 expression in human chondrocytes. Am. J. Transl. Res. 2015, 7, 2000–2006.
  117. Vonk, L.A.; Kragten, A.H.M.; Dhert, W.J.A.; Saris, D.B.F.; Creemers, L.B. Overexpression of hsa-miR-148a promotes cartilage production and inhibits cartilage degradation by osteoarthritic chondrocytes. Osteoarthr. Cartil. 2014, 22, 145–153.
  118. Tardif, G.; Hum, D.; Pelletier, J.P.; Duval, N.; Martel-Pelletier, J. Regulation of the IGFBP-5 and MMP-13 genes by the microRNAs miR-140 and miR-27a in human osteoarthritic chondrocytes. BMC Musculoskelet. Disord. 2009, 10.
  119. Li, X.; Zhen, Z.; Tang, G.; Zheng, C.; Yang, G. MiR-29a and MiR-140 protect chondrocytes against the anti-proliferation and cell matrix signaling changes by IL-1β. Mol. Cells 2016, 39, 103–110.
  120. Song, J.; Jin, E.H.; Kim, D.; Kim, K.Y.; Chun, C.H.; Jin, E.J. MicroRNA-222 regulates MMP-13 via targeting HDAC-4 during osteoarthritis pathogenesis. BBA Clin. 2015, 3, 79–89.
  121. Song, J.; Kim, D.; Lee, C.H.; Lee, M.S.; Chun, C.H.; Jin, E.J. MicroRNA-488 regulates zinc transporter SLC39A8/ZIP8 during pathogenesis of osteoarthritis. J. Biomed. Sci. 2013, 20.
  122. Akhtar, N.; Makki, M.S.; Haqqi, T.M. MicroRNA-602 and microRNA-608 regulate sonic hedgehog expression via target sites in the coding region in human chondrocytes. Arthritis Rheumatol. 2015, 67, 423–434.
  123. Philipot, D.; Guérit, D.; Platano, D.; Chuchana, P.; Olivotto, E.; Espinoza, F.; Dorandeu, A.; Pers, Y.M.; Piette, J.; Borzi, R.M.; et al. P16INK4a and its regulator miR-24 link senescence and chondrocyte terminal differentiation-associated matrix remodeling in osteoarthritis. Arthritis Res. Ther. 2014, 16.
  124. Rasheed, Z.; Rasheed, N.; Abdulmonem, W.A.; Khan, M.I. MicroRNA-125b-5p regulates IL-1β induced inflammatory genes via targeting TRAF6-mediated MAPKs and NF-κB signaling in human osteoarthritic chondrocytes. Sci. Rep. 2019, 9.
  125. Kostopoulou, F.; Malizos, K.N.; Papathanasiou, I.; Tsezou, A. MicroRNA-33a regulates cholesterol synthesis and cholesterol efflux-related genes in osteoarthritic chondrocytes. Arthritis Res. Ther. 2015, 17.
  126. Song, J.; Lee, M.; Kim, D.; Han, J.; Chun, C.H.; Jin, E.J. MicroRNA-181b regulates articular chondrocytes differentiation and cartilage integrity. Biochem. Biophys. Res. Commun. 2013, 431, 210–214.
  127. Yang, B.; Kang, X.; Xing, Y.; Dou, C.; Kang, F.; Li, J.; Quan, Y.; Dong, S. Effect of microRNA-145 on IL-1β-induced cartilage degradation in human chondrocytes. FEBS Lett. 2014, 588, 2344–2352.
  128. Li, L.; Jia, J.; Liu, X.; Yang, S.; Ye, S.; Yang, W.; Zhang, Y. MicroRNA-16-5p Controls Development of Osteoarthritis by Targeting SMAD3 in Chondrocytes. Curr. Pharm. Des. 2015, 21, 5160–5167.
  129. Qi, Y.; Ma, N.; Yan, F.; Yu, Z.; Wu, G.; Qiao, Y.; Han, D.; Xiang, Y.; Li, F.; Wang, W.; et al. The expression of intronic miRNAs, miR-483 and miR-483*, and their host gene, Igf2, in murine osteoarthritis cartilage. Int. J. Biol. Macromol. 2013, 61, 43–49.
  130. McManus, M.T.; Sharp, P.A. Gene silencing in mammals by small interfering RNAs. Nat. Rev. Genet. 2002, 3, 737–747.
  131. Hoshi, H.; Akagi, R.; Yamaguchi, S.; Muramatsu, Y.; Akatsu, Y.; Yamamoto, Y.; Sasaki, T.; Takahashi, K.; Sasho, T. Effect of inhibiting MMP13 and ADAMTS5 by intra-articular injection of small interfering RNA in a surgically induced osteoarthritis model of mice. Cell Tissue Res. 2017, 368, 379–387.
  132. Akagi, R.; Sasho, T.; Saito, M.; Endo, J.; Yamaguchi, S.; Muramatsu, Y.; Mukoyama, S.; Akatsu, Y.; Katsuragi, J.; Fukawa, T.; et al. Effective knock down of matrix metalloproteinase-13 by an intra-articular injection of small interfering RNA (siRNA) in a murine surgically-induced osteoarthritis model. J. Orthop. Res. 2014, 32, 1175–1180.
  133. Hoshi, H.; Sasho, T.; Akagi, R.; Muramatsu, Y.; Mukoyama, S.; Akatsu, Y.; Fukawa, T.; Katsuragi, J.; Endo, J.; Yamamoto, Y. Effective knock down of MMP13 and ADAMTS5 by intra-articular injection of small interference RNA (siRNA) in a surgically induced osteoarthritis model of mice. Osteoarthr. Cartil. 2014, 22, S372–S373.
  134. Mercer, T.R.; Dinger, M.E.; Mattick, J.S. Long non-coding RNAs: Insights into functions. Nat. Rev. Genet. 2009, 10, 155–159.
  135. Fu, M.; Huang, G.; Zhang, Z.; Liu, J.; Zhang, Z.; Huang, Z.; Yu, B.; Meng, F. Expression profile of long noncoding RNAs in cartilage from knee osteoarthritis patients. Osteoarthr. Cartil. 2015, 23, 423–432.
  136. Liu, Q.; Zhang, X.; Dai, L.; Hu, X.; Zhu, J.; Li, L.; Zhou, C.; Ao, Y. Long noncoding RNA related to cartilage injury promotes chondrocyte extracellular matrix degradation in osteoarthritis. Arthritis Rheumatol. 2014, 66, 969–978.
  137. Song, J.; Ahn, C.; Chun, C.H.; Jin, E.J. A long non-coding RNA, GAS5, plays a critical role in the regulation of miR-21 during osteoarthritis. J. Orthop. Res. 2014, 32, 1628–1635.
  138. Wang, G.; Bu, X.; Zhang, Y.; Zhao, X.; Kong, Y.; Ma, L.; Niu, S.; Wu, B.; Meng, C. LncRNA-UCA1 enhances MMP-13 expression by inhibiting miR- 204-5p in human chondrocytes. Oncotarget 2017, 8, 91281–91290.
  139. Seda Yar Saglam, A.; Alp, E.; Ilke Onen, H. Circular RNAs and Its Biological Functions in Health and Disease. In Gene Expression and Phenotypic Traits; IntechOpen: London, UK, 2020.
  140. Hansen, T.B.; Jensen, T.I.; Clausen, B.H.; Bramsen, J.B.; Finsen, B.; Damgaard, C.K.; Kjems, J. Natural RNA circles function as efficient microRNA sponges. Nature 2013, 495, 384–388.
  141. Liu, Q.; Zhang, X.; Hu, X.; Dai, L.; Fu, X.; Zhang, J.; Ao, Y. Circular RNA Related to the Chondrocyte ECM Regulates MMP13 Expression by Functioning as a MiR-136 “Sponge” in Human Cartilage Degradation. Sci. Rep. 2016, 6.
  142. Gu, H.; Jiao, Y.; Yu, X.; Li, X.; Wang, W.; Ding, L.; Liu, L. Resveratrol inhibits the IL-1β-induced expression ofMMP-13 and IL-6 in human articular chondrocytes viaTLR4/MyD88-dependent and-independent signaling cascades. Int. J. Mol. Med. 2017, 39, 734–740.
  143. Yang, Q.; Wu, S.; Mao, X.; Wang, W.; Tai, H. Inhibition effect of curcumin on TNF-α and MMP-13 expression induced by advanced glycation end products in chondrocytes. Pharmacology 2013, 91, 77–85.
  144. Rasheed, Z.; Anbazhagan, A.N.; Akhtar, N.; Ramamurthy, S.; Voss, F.R.; Haqqi, T.M. Green tea polyphenol epigallocatechin-3-gallate inhibits advanced glycation end product-induced expression of tumor necrosis factor-α and matrix metalloproteinase-13 in human chondrocytes. Arthritis Res. Ther. 2009, 11.
  145. Janusz, M.J.; Hookfin, E.B.; Heitmeyer, S.A.; Woessner, J.F.; Freemont, A.J.; Hoyland, J.A.; Brown, K.K.; Hsieh, L.C.; Almstead, N.G.; De, B.; et al. Moderation of iodoacetate-induced experimental osteoarthritis in rats by matrix metalloproteinase inhibitors. Osteoarthr. Cartil. 2001, 9, 751–760.
  146. Fingleton, B. Matrix Metalloproteinases as Valid Clinical Target. Curr. Pharm. Des. 2006, 13, 333–346.
  147. Johnson, A.R.; Pavlovsky, A.G.; Ortwine, D.F.; Prior, F.; Man, C.F.; Bornemeier, D.A.; Banotai, C.A.; Mueller, W.T.; McConnell, P.; Yan, C.; 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. 2007, 282, 27781–27791.
  148. Nuti, E.; Casalini, F.; Avramova, S.I.; Santamaria, S.; Cercignani, G.; Marinelli, L.; La Pietra, V.; Novellino, E.; Orlandini, E.; Nencetti, S.; et al. N-O-isopropyl sulfonamido-based hydroxamates: Design, synthesis and biological evaluation of selective matrix metalloproteinase-13 inhibitors as potential therapeutic agents for osteoarthritis. J. Med. Chem. 2009, 52, 4757–4773.
  149. Monovich, L.G.; Tommasi, R.A.; Fujimoto, R.A.; Blancuzzi, V.; Clark, K.; Cornell, W.D.; Doti, R.; Doughty, J.; Fang, J.; Farley, D.; et al. Discovery of potent, selective, and orally active carboxylic acid based inhibitors of matrix metalloproteinase-13. J. Med. Chem. 2009, 52, 3523–3538.
  150. Nara, H.; Sato, K.; Kaieda, A.; Oki, H.; Kuno, H.; Santou, T.; Kanzaki, N.; Terauchi, J.; Uchikawa, O.; Kori, M. Design, synthesis, and biological activity of novel, potent, and highly selective fused pyrimidine-2-carboxamide-4-one-based matrix metalloproteinase (MMP)-13 zinc-binding inhibitors. Bioorg. Med. Chem. 2016, 24, 6149–6165.
  151. Nara, H.; Kaieda, A.; Sato, K.; Naito, T.; Mototani, H.; Oki, H.; Yamamoto, Y.; Kuno, H.; Santou, T.; Kanzaki, N.; et al. Discovery of novel, highly potent, and selective matrix metalloproteinase (MMP)-13 inhibitors with a 1,2,4-triazol-3-yl moiety as a zinc binding group using a structure-based design approach. J. Med. Chem. 2017, 60, 608–626.
  152. De Savi, C.; Morley, A.D.; Ting, A.; Nash, I.; Karabelas, K.; Wood, C.M.; James, M.; Norris, S.J.; Karoutchi, G.; Rankine, N.; et al. Selective non zinc binding inhibitors of MMP13. Bioorg. Med. Chem. Lett. 2011, 21, 4215–4219.
  153. Jie, J.L.; Nahra, J.; Johnson, A.R.; Bunker, A.; O’Brien, P.; Yue, W.S.; Ortwine, D.F.; Man, C.F.; Baragi, V.; Kilgore, K.; et al. Quinazolinones and pyrido[3,4-d]pyrimidin-4-ones as orally active and specific matrix metalloproteinase-13 inhibitors for the treatment of osteoarthritis. J. Med. Chem. 2008, 51, 835–841.
  154. Schnute, M.E.; O’Brien, P.M.; Nahra, J.; Morris, M.; Howard Roark, W.; Hanau, C.E.; Ruminski, P.G.; Scholten, J.A.; Fletcher, T.R.; Hamper, B.C.; et al. Discovery of (pyridin-4-yl)-2H-tetrazole as a novel scaffold to identify highly selective matrix metalloproteinase-13 inhibitors for the treatment of osteoarthritis. Bioorg. Med. Chem. Lett. 2010, 20, 576–580.
  155. Gege, C.; Bao, B.; Bluhm, H.; Boer, J.; Gallagher, B.M.; Korniski, B.; Powers, T.S.; Steeneck, C.; Taveras, A.G.; Baragi, V.M. Discovery and evaluation of a non-Zn chelating, selective matrix metalloproteinase 13 (MMP-13) inhibitor for potential intra-articular treatment of osteoarthritis. J. Med. Chem. 2012, 55, 709–716.
  156. Settle, S.; Vickery, L.; Nemirovskiy, O.; Vidmar, T.; Bendele, A.; Messing, D.; Ruminski, P.; Schnute, M.; Sunyer, T. Cartilage degradation biomarkers predict efficacy of a novel, highly selective matrix metalloproteinase 13 inhibitor in a dog model of osteoarthritis: Confirmation by multivariate analysis that modulation of type II collagen and aggrecan degradation pepti. Arthritis Rheum. 2010, 62, 3006–3015.
  157. Ruminski, P.G.; Massa, M.; Strohbach, J.; Hanau, C.E.; Schmidt, M.; Scholten, J.A.; Fletcher, T.R.; Hamper, B.C.; Carroll, J.N.; Shieh, H.S.; et al. Discovery of N-(4-Fluoro-3-methoxybenzyl)-6-(2-(((2S,5R)-5-(hydroxymethyl)-1,4-dioxan-2-yl)methyl)-2H-tetrazol-5-yl)-2-methylpyrimidine-4-carboxamide. A Highly Selective and Orally Bioavailable Matrix Metalloproteinase-13 Inhibitor for the Potential Treat. J. Med. Chem. 2016, 59, 313–327.
  158. Zheng, S.; Hunter, D.J.; Xu, J.; Ding, C. Monoclonal antibodies for the treatment of osteoarthritis. Expert Opin. Biol. Ther. 2016, 16, 1529–1540.
  159. Naito, S.; Takahashi, T.; Onoda, J.; Yamauchi, A.; Kawai, T.; Kishino, J.; Yamane, S.; Fujii, I.; Fukui, N.; Numata, Y. Development of a neutralizing antibody specific for the active form of matrix metalloproteinase-13. Biochemistry 2012, 51, 8877–8884.
  160. Amar, S.; Fields, G.B. Potential clinical implications of recent matrix metalloproteinase inhibitor design strategies. Expert Rev. Proteomics 2015, 12, 445–447.
More