DOI QR코드

DOI QR Code

Vitamin D Attenuates Pain and Cartilage Destruction in OA Animals via Enhancing Autophagic Flux and Attenuating Inflammatory Cell Death

  • JooYeon Jhun (Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea) ;
  • Jin Seok Woo (Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea) ;
  • Ji Ye Kwon (Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea) ;
  • Hyun Sik Na (Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea) ;
  • Keun-Hyung Cho (Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea) ;
  • Seon Ae Kim (Department of Orthopedic Surgery, College of Medicine, The Catholic University of Korea) ;
  • Seok Jung Kim (Department of Orthopedic Surgery, College of Medicine, The Catholic University of Korea) ;
  • Su-Jin Moon (Division of Rheumatology, Department of Internal Medicine, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea) ;
  • Sung-Hwan Park (Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea) ;
  • Mi-La Cho (Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea)
  • Received : 2021.12.14
  • Accepted : 2022.04.10
  • Published : 2022.08.31

Abstract

Osteoarthritis (OA) is the most common form of arthritis associated with ageing. Vitamin D has diverse biological effect on bone and cartilage, and observational studies have suggested it potential benefit in OA progression and inflammation process. However, the effect of vitamin D on OA is still contradictory. Here, we investigated the therapeutic potential of vitamin D in OA. Six-week-old male Wistar rats were injected with monosodium iodoacetate (MIA) to induce OA. Pain severity, cartilage destruction, and inflammation were measured in MIA-induced OA rats. Autophagy activity and mitochondrial function were also measured. Vitamin-D (1,25(OH)2D3) and celecoxib were used to treat MIA-induced OA rats and OA chondrocytes. Oral supplementation of vitamin D resulted in significant attenuations in OA pain, inflammation, and cartilage destruction. Interestingly, the expressions of MMP-13, IL-1β, and MCP-1 in synovial tissues were remarkably attenuated by vitamin D treatment, suggesting its potential to attenuate synovitis in OA. Vitamin D treatment in OA chondrocytes resulted in autophagy induction in human OA chondrocytes and increased expression of TFEB, but not LC3B, caspase-1 and -3, in inflamed synovium. Vitamin D and celecoxib showed a synergistic effect on antinociceptive and chondroprotective properties in vivo. Vitamin D showed the chondroprotective and antinociceptive property in OA rats. Autophagy induction by vitamin D treatment may be a promising treatment strategy in OA patients especially presenting vitamin D deficiency. Autophagy promoting strategy may attenuate OA progression through protecting cells from damage and inflammatory cell death.

Keywords

Acknowledgement

This research was supported by Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Education (grant number NRF-2015R1D1A1A01057065 and 2020R1I1A1A01072520).

References

  1. Martel-Pelletier J, Barr AJ, Cicuttini FM, Conaghan PG, Cooper C, Goldring MB, Goldring SR, Jones G, Teichtahl AJ, Pelletier JP. Osteoarthritis. Nat Rev Dis Primers 2016;2:16072.
  2. Bowden JL, Hunter DJ, Deveza LA, Duong V, Dziedzic KS, Allen KD, Chan PK, Eyles JP. Core and adjunctive interventions for osteoarthritis: efficacy and models for implementation. Nat Rev Rheumatol 2020;16:434-447. https://doi.org/10.1038/s41584-020-0447-8
  3. Moon SJ, Jeong JH, Jhun JY, Yang EJ, Min JK, Choi JY, Cho ML. Ursodeoxycholic acid ameliorates pain severity and cartilage degeneration in monosodium iodoacetate-induced osteoarthritis in rats. Immune Netw 2014;14:45-53. https://doi.org/10.4110/in.2014.14.1.45
  4. Carames B, Taniguchi N, Otsuki S, Blanco FJ, Lotz M. Autophagy is a protective mechanism in normal cartilage, and its aging-related loss is linked with cell death and osteoarthritis. Arthritis Rheum 2010;62:791-801.
  5. Sellam J, Berenbaum F. The role of synovitis in pathophysiology and clinical symptoms of osteoarthritis. Nat Rev Rheumatol 2010;6:625-635. https://doi.org/10.1038/nrrheum.2010.159
  6. Prieto-Alhambra D, Judge A, Javaid MK, Cooper C, Diez-Perez A, Arden NK. Incidence and risk factors for clinically diagnosed knee, hip and hand osteoarthritis: influences of age, gender and osteoarthritis affecting other joints. Ann Rheum Dis 2014;73:1659-1664. https://doi.org/10.1136/annrheumdis-2013-203355
  7. Zheng H, Chen C. Body mass index and risk of knee osteoarthritis: systematic review and meta-analysis of prospective studies. BMJ Open 2015;5:e007568.
  8. Slauterbeck JR, Kousa P, Clifton BC, Naud S, Tourville TW, Johnson RJ, Beynnon BD. Geographic mapping of meniscus and cartilage lesions associated with anterior cruciate ligament injuries. J Bone Joint Surg Am 2009;91:2094-2103. https://doi.org/10.2106/JBJS.H.00888
  9. Lane NE, Gore LR, Cummings SR, Hochberg MC, Scott JC, Williams EN, Nevitt MC. Serum vitamin D levels and incident changes of radiographic hip osteoarthritis: a longitudinal study. Study of osteoporotic fractures research group. Arthritis Rheum 1999;42:854-860. https://doi.org/10.1002/1529-0131(199905)42:5<854::AID-ANR3>3.0.CO;2-I
  10. Chang Z, Huo L, Li P, Wu Y, Zhang P. Ascorbic acid provides protection for human chondrocytes against oxidative stress. Mol Med Rep 2015;12:7086-7092. https://doi.org/10.3892/mmr.2015.4231
  11. McAlindon TE, Felson DT, Zhang Y, Hannan MT, Aliabadi P, Weissman B, Rush D, Wilson PW, Jacques P. Relation of dietary intake and serum levels of vitamin D to progression of osteoarthritis of the knee among participants in the Framingham Study. Ann Intern Med 1996;125:353-359. https://doi.org/10.7326/0003-4819-125-5-199609010-00001
  12. Bergink AP, Uitterlinden AG, Van Leeuwen JP, Buurman CJ, Hofman A, Verhaar JA, Pols HA. Vitamin D status, bone mineral density, and the development of radiographic osteoarthritis of the knee: the Rotterdam Study. J Clin Rheumatol 2009;15:230-237. https://doi.org/10.1097/RHU.0b013e3181b08f20
  13. Heidari B, Heidari P, Hajian-Tilaki K. Association between serum vitamin D deficiency and knee osteoarthritis. Int Orthop 2011;35:1627-1631. https://doi.org/10.1007/s00264-010-1186-2
  14. Felson DT, Niu J, Clancy M, Aliabadi P, Sack B, Guermazi A, Hunter DJ, Amin S, Rogers G, Booth SL. Low levels of vitamin D and worsening of knee osteoarthritis: results of two longitudinal studies. Arthritis Rheum 2007;56:129-136. https://doi.org/10.1002/art.22292
  15. McAlindon T, LaValley M, Schneider E, Nuite M, Lee JY, Price LL, Lo G, Dawson-Hughes B. Effect of vitamin D supplementation on progression of knee pain and cartilage volume loss in patients with symptomatic osteoarthritis: a randomized controlled trial. JAMA 2013;309:155-162. https://doi.org/10.1001/jama.2012.164487
  16. Jin X, Jones G, Cicuttini F, Wluka A, Zhu Z, Han W, Antony B, Wang X, Winzenberg T, Blizzard L, et al. Effect of vitamin D supplementation on tibial cartilage volume and knee pain among patients with symptomatic knee osteoarthritis: a randomized clinical trial. JAMA 2016;315:1005-1013. https://doi.org/10.1001/jama.2016.1961
  17. Perry TA, Parkes MJ, Hodgson R, Felson DT, O'Neill TW, Arden NK. Effect of vitamin D supplementation on synovial tissue volume and subchondral bone marrow lesion volume in symptomatic knee osteoarthritis. BMC Musculoskelet Disord 2019;20:76.
  18. Agergaard J, Trostrup J, Uth J, Iversen JV, Boesen A, Andersen JL, Schjerling P, Langberg H. Does vitamin-D intake during resistance training improve the skeletal muscle hypertrophic and strength response in young and elderly men? - A randomized controlled trial. Nutr Metab (Lond) 2015;12:32.
  19. Granic A, Hill TR, Davies K, Jagger C, Adamson A, Siervo M, Kirkwood TB, Mathers JC, Sayer AA. Vitamin D status, muscle strength and physical performance decline in very old adults: a prospective study. Nutrients 2017;9:379.
  20. Chung C, Silwal P, Kim I, Modlin RL, Jo EK. Vitamin D-cathelicidin axis: at the crossroads between protective immunity and pathological inflammation during infection. Immune Netw 2020;20:e12.
  21. Pojednic RM, Ceglia L. The emerging biomolecular role of vitamin D in skeletal muscle. Exerc Sport Sci Rev 2014;42:76-81. https://doi.org/10.1249/JES.0000000000000013
  22. Ruiz-Romero C, Calamia V, Mateos J, Carreira V, Martinez-Gomariz M, Fernandez M, Blanco FJ. Mitochondrial dysregulation of osteoarthritic human articular chondrocytes analyzed by proteomics: a decrease in mitochondrial superoxide dismutase points to a redox imbalance. Mol Cell Proteomics 2009;8:172-189. https://doi.org/10.1074/mcp.M800292-MCP200
  23. Dave M, Attur M, Palmer G, Al-Mussawir HE, Kennish L, Patel J, Abramson SB. The antioxidant resveratrol protects against chondrocyte apoptosis via effects on mitochondrial polarization and ATP production. Arthritis Rheum 2008;58:2786-2797. https://doi.org/10.1002/art.23799
  24. Dzik KP, Skrobot W, Kaczor KB, Flis DJ, Karnia MJ, Libionka W, Antosiewicz J, Kloc W, Kaczor JJ. Vitamin D deficiency is associated with muscle atrophy and reduced mitochondrial function in patients with chronic low back pain. Oxid Med Cell Longev 2019;2019:6835341.
  25. Stio M, Lunghi B, Iantomasi T, Vincenzini MT, Treves C. Effect of vitamin D deficiency and 1,25-dihydroxyvitamin D3 on rat heart metabolism. J Mol Cell Cardiol 1994;26:1421-1428. https://doi.org/10.1006/jmcc.1994.1161
  26. Ashcroft SP, Fletcher G, Philp AM, Jenkinson C, Das S, Hansbro PM, Atherton PJ, Philp A. Diet-induced vitamin D deficiency reduces skeletal muscle mitochondrial respiration. J Endocrinol 2021;249:113-124. https://doi.org/10.1530/JOE-20-0233
  27. Blajszczak CC, Nonn L. Vitamin D regulates prostate cell metabolism via genomic and non-genomic mitochondrial redox-dependent mechanisms. J Steroid Biochem Mol Biol 2019;195:105484.
  28. MacLaughlin J, Holick MF. Aging decreases the capacity of human skin to produce vitamin D3. J Clin Invest 1985;76:1536-1538. https://doi.org/10.1172/JCI112134
  29. Wang Y, Zhu J, DeLuca HF. Identification of the vitamin D receptor in osteoblasts and chondrocytes but not osteoclasts in mouse bone. J Bone Miner Res 2014;29:685-692. https://doi.org/10.1002/jbmr.2081
  30. Tetlow LC, Woolley DE. Expression of vitamin D receptors and matrix metalloproteinases in osteoarthritic cartilage and human articular chondrocytes in vitro. Osteoarthritis Cartilage 2001;9:423-431. https://doi.org/10.1053/joca.2000.0408
  31. Mizushima N, Komatsu M. Autophagy: renovation of cells and tissues. Cell 2011;147:728-741. https://doi.org/10.1016/j.cell.2011.10.026
  32. Marycz K, Weiss C, Smieszek A, Kornicka K. Evaluation of oxidative stress and mitophagy during adipogenic differentiation of adipose-derived stem cells isolated from equine metabolic syndrome (EMS) horses. Stem Cells Int 2018;2018:5340756.
  33. Son J, Kim MJ, Lee JS, Kim JY, Chun E, Lee KY. Hepatitis B virus X protein promotes liver cancer progression through autophagy induction in response to TLR4 stimulation. Immune Netw 2021;21:e37.
  34. Rockel JS, Kapoor M. Autophagy: controlling cell fate in rheumatic diseases. Nat Rev Rheumatol 2016;12:517-531. https://doi.org/10.1038/nrrheum.2016.92
  35. Lopez de Figueroa P, Lotz MK, Blanco FJ, Carames B. Autophagy activation and protection from mitochondrial dysfunction in human chondrocytes. Arthritis Rheumatol 2015;67:966-976. https://doi.org/10.1002/art.39025
  36. Feng K, Chen H, Xu C. Chondro-protective effects of celastrol on osteoarthritis through autophagy activation and NF-κB signaling pathway inhibition. Inflamm Res 2020;69:385-400. https://doi.org/10.1007/s00011-020-01327-z
  37. Yang H, Wen Y, Zhang M, Liu Q, Zhang H, Zhang J, Lu L, Ye T, Bai X, Xiao G, et al. MTORC1 coordinates the autophagy and apoptosis signaling in articular chondrocytes in osteoarthritic temporomandibular joint. Autophagy 2020;16:271-288. https://doi.org/10.1080/15548627.2019.1606647
  38. Wang C, Yao Z, Zhang Y, Yang Y, Liu J, Shi Y, Zhang C. Metformin mitigates cartilage degradation by activating AMPK/SIRT1-mediated autophagy in a mouse osteoarthritis model. Front Pharmacol 2020;11:1114.
  39. Galluzzi L, Bravo-San Pedro JM, Levine B, Green DR, Kroemer G. Pharmacological modulation of autophagy: therapeutic potential and persisting obstacles. Nat Rev Drug Discov 2017;16:487-511. https://doi.org/10.1038/nrd.2017.22
  40. Carames B, Taniguchi N, Seino D, Blanco FJ, D'Lima D, Lotz M. Mechanical injury suppresses autophagy regulators and pharmacologic activation of autophagy results in chondroprotection. Arthritis Rheum 2012;64:1182-1192. https://doi.org/10.1002/art.33444
  41. Sasaki H, Takayama K, Matsushita T, Ishida K, Kubo S, Matsumoto T, Fujita N, Oka S, Kurosaka M, Kuroda R. Autophagy modulates osteoarthritis-related gene expression in human chondrocytes. Arthritis Rheum 2012;64:1920-1928. https://doi.org/10.1002/art.34323
  42. Lee J, Hong YS, Jeong JH, Yang EJ, Jhun JY, Park MK, Jung YO, Min JK, Kim HY, Park SH, et al. Coenzyme Q10 ameliorates pain and cartilage degradation in a rat model of osteoarthritis by regulating nitric oxide and inflammatory cytokines. PLoS One 2013;8:e69362.
  43. Lee SY, Lee SH, Na HS, Kwon JY, Kim GY, Jung K, Cho KH, Kim SA, Go EJ, Park MJ, et al. The therapeutic effect of STAT3 signaling-suppressed MSC on pain and articular cartilage damage in a rat model of monosodium iodoacetate-induced osteoarthritis. Front Immunol 2018;9:2881.
  44. Kwon JY, Lee SH, Na HS, Jung K, Choi J, Cho KH, Lee CY, Kim SJ, Park SH, Shin DY, et al. Kartogenin inhibits pain behavior, chondrocyte inflammation, and attenuates osteoarthritis progression in mice through induction of IL-10. Sci Rep 2018;8:13832.
  45. Pauli C, Whiteside R, Heras FL, Nesic D, Koziol J, Grogan SP, Matyas J, Pritzker KP, D'Lima DD, Lotz MK. Comparison of cartilage histopathology assessment systems on human knee joints at all stages of osteoarthritis development. Osteoarthritis Cartilage 2012;20:476-485. https://doi.org/10.1016/j.joca.2011.12.018
  46. Pritzker KP, Gay S, Jimenez SA, Ostergaard K, Pelletier JP, Revell PA, Salter D, van den Berg WB. Osteoarthritis cartilage histopathology: grading and staging. Osteoarthritis Cartilage 2006;14:13-29. https://doi.org/10.1016/j.joca.2005.07.014
  47. Davidson RK, Waters JG, Kevorkian L, Darrah C, Cooper A, Donell ST, Clark IM. Expression profiling of metalloproteinases and their inhibitors in synovium and cartilage. Arthritis Res Ther 2006;8:R124.
  48. Xu YK, Ke Y, Wang B, Lin JH. The role of MCP-1-CCR2 ligand-receptor axis in chondrocyte degradation and disease progress in knee osteoarthritis. Biol Res 2015;48:64.
  49. Raghu H, Lepus CM, Wang Q, Wong HH, Lingampalli N, Oliviero F, Punzi L, Giori NJ, Goodman SB, Chu CR, et al. CCL2/CCR2, but not CCL5/CCR5, mediates monocyte recruitment, inflammation and cartilage destruction in osteoarthritis. Ann Rheum Dis 2017;76:914-922. https://doi.org/10.1136/annrheumdis-2016-210426
  50. Blanco FJ, Rego I, Ruiz-Romero C. The role of mitochondria in osteoarthritis. Nat Rev Rheumatol 2011;7:161-169. https://doi.org/10.1038/nrrheum.2010.213
  51. Rambold AS, Lippincott-Schwartz J. Mechanisms of mitochondria and autophagy crosstalk. Cell Cycle 2011;10:4032-4038. https://doi.org/10.4161/cc.10.23.18384
  52. Tang Q, Zheng G, Feng Z, Chen Y, Lou Y, Wang C, Zhang X, Zhang Y, Xu H, Shang P, et al. Trehalose ameliorates oxidative stress-mediated mitochondrial dysfunction and ER stress via selective autophagy stimulation and autophagic flux restoration in osteoarthritis development. Cell Death Dis 2017;8:e3081.
  53. Ansari MY, Ball HC, Wase SJ, Novak K, Haqqi TM. Lysosomal dysfunction in osteoarthritis and aged cartilage triggers apoptosis in chondrocytes through BAX mediated release of Cytochrome c. Osteoarthritis Cartilage 2021;29:100-112. https://doi.org/10.1016/j.joca.2020.08.014
  54. Nakamura S, Shigeyama S, Minami S, Shima T, Akayama S, Matsuda T, Esposito A, Napolitano G, Kuma A, Namba-Hamano T, et al. LC3 lipidation is essential for TFEB activation during the lysosomal damage response to kidney injury. Nat Cell Biol 2020;22:1252-1263. https://doi.org/10.1038/s41556-020-00583-9
  55. Runwal G, Stamatakou E, Siddiqi FH, Puri C, Zhu Y, Rubinsztein DC. LC3-positive structures are prominent in autophagy-deficient cells. Sci Rep 2019;9:10147.
  56. Evans CH, Kraus VB, Setton LA. Progress in intra-articular therapy. Nat Rev Rheumatol 2014;10:11-22. https://doi.org/10.1038/nrrheum.2013.159
  57. Liu Q, Niu J, Li H, Ke Y, Li R, Zhang Y, Lin J. Knee symptomatic osteoarthritis, walking disability, NSAIDs use and all-cause mortality: population-based Wuchuan Osteoarthritis Study. Sci Rep 2017;7:3309.
  58. Fu J, Li S, Feng R, Ma H, Sabeh F, Roodman GD, Wang J, Robinson S, Guo XE, Lund T, et al. Multiple myeloma-derived MMP-13 mediates osteoclast fusogenesis and osteolytic disease. J Clin Invest 2016;126:1759-1772. https://doi.org/10.1172/JCI80276
  59. Park MJ, Moon SJ, Baek JA, Lee EJ, Jung KA, Kim EK, Kim DS, Lee JH, Kwok SK, Min JK, et al. Metformin augments anti-inflammatory and chondroprotective properties of mesenchymal stem cells in experimental osteoarthritis. J Immunol 2019;203:127-136. https://doi.org/10.4049/jimmunol.1800006
  60. Sutton CE, Lalor SJ, Sweeney CM, Brereton CF, Lavelle EC, Mills KH. Interleukin-1 and IL-23 induce innate IL-17 production from γδ T cells, amplifying Th17 responses and autoimmunity. Immunity 2009;31:331-341. https://doi.org/10.1016/j.immuni.2009.08.001
  61. Li Z, Cheng J, Liu J. Baicalin protects human OA chondrocytes against IL-1β-induced apoptosis and ECM degradation by activating autophagy via miR-766-3p/AIFM1 axis. Drug Des Devel Ther 2020;14:2645-2655. https://doi.org/10.2147/DDDT.S255823
  62. Kong C, Wang C, Shi Y, Yan L, Xu J, Qi W. Active vitamin D activates chondrocyte autophagy to reduce osteoarthritis via mediating the AMPK-mTOR signaling pathway. Biochem Cell Biol 2020;98:434-442. https://doi.org/10.1139/bcb-2019-0333
  63. Wang J, Lian H, Zhao Y, Kauss MA, Spindel S. Vitamin D3 induces autophagy of human myeloid leukemia cells. J Biol Chem 2008;283:25596-25605. https://doi.org/10.1074/jbc.M801716200
  64. Lisse TS, Hewison M. Vitamin D: a new player in the world of mTOR signaling. Cell Cycle 2011;10:1888-1889. https://doi.org/10.4161/cc.10.12.15620
  65. Chen YC, Sung HC, Chuang TY, Lai TC, Lee TL, Lee CW, Lee IT, Chen YL. Vitamin D3 decreases TNF-α-induced inflammation in lung epithelial cells through a reduction in mitochondrial fission and mitophagy. Cell Biol Toxicol 2021;1-24.
  66. Lee TL, Lee MH, Chen YC, Lee YC, Lai TC, Lin HY, Hsu LF, Sung HC, Lee CW, Chen YL. Vitamin D attenuates ischemia/reperfusion-induced cardiac injury by reducing mitochondrial fission and mitophagy. Front Pharmacol 2020;11:604700. 
  67. Minisola S, Ferrone F, Danese V, Cecchetti V, Pepe J, Cipriani C, Colangelo L. Controversies surrounding vitamin D: focus on supplementation and cancer. Int J Environ Res Public Health 2019;16:189.
  68. Lewis JR, Sim M, Daly RM. The vitamin D and calcium controversy: an update. Curr Opin Rheumatol 2019;31:91-97. https://doi.org/10.1097/BOR.0000000000000584
  69. Glendenning P, Inderjeeth CA. Controversy and consensus regarding vitamin D: recent methodological changes and the risks and benefits of vitamin D supplementation. Crit Rev Clin Lab Sci 2016;53:13-28. https://doi.org/10.3109/10408363.2015.1074157
  70. Jin X, Antony B, Wang X, Persson MS, McAlindon T, Arden NK, Srivastava S, Srivastava R, Van Middelkoop M, Bierma-Zeinstra SM, et al. Effect of vitamin D supplementation on pain and physical function in patients with knee osteoarthritis (OA): an OA Trial Bank protocol for a systematic review and individual patient data (IPD) meta-analysis. BMJ Open 2020;10:e035302.
  71. Tu L, Zheng S, Cicuttini F, Jin X, Han W, Zhu Z, Antony B, Winzenberg T, Jones G, Gu J, et al. Effects of vitamin D supplementation on disabling foot pain in patients with symptomatic knee osteoarthritis. Arthritis Care Res (Hoboken) 2021;73:781-7. https://doi.org/10.1002/acr.24371