Online first
Original paper
Published online: 2024-08-14

open access

Page views 6
Article views/downloads 7
Get Citation

Connect on Social Media

Connect on Social Media

Platelet-rich plasma ameliorates cartilage degradation in rat models of osteoarthritis via the OPG/RANKL/RANK system

Qun Wu1, Xianbao Yao1, Nan Shan1, Yi Cai1, Yongzhi Fan1

Abstract

Introduction. Osteoarthritis (OA) is one of the most common degenerative joint diseases in the elderly, which is featured by the degradation of articular cartilage. Recently, platelet-rich plasma (PRP) injection into the affected joint has become one biological therapy for OA treatment. The OPG/RANKL/RANK signalling has been reported to mediate OA progression. Our study aimed to confirm whether PRP injection retards OA development through the regulation of the OPG/RANKL/RANK system.

Material and methods. The OA rat models were induced by medial menisci resection combined with anterior cruciate ligament transection. Four weeks after surgery, OA-induced rats received intra-articular injection with 50 μL PRP once a week for 6 weeks. Rats were euthanised one week after the 6th injection. Rat knee joints were subjected to histopathological examination by haematoxylin-eosin (H&E) and safranin O staining. Osteoprotegerin (OPG), receptor activator of nuclear factor kappa B (RANK), and RANK ligand (RANKL) in the articular cartilage of rats were tested through immunofluorescence staining and western blotting. Serum interleukin-1β (IL-1β) and interleukin-6 (IL-6) levels were measured by enzyme-linked immunosorbent assay (ELISA). Matrix metalloproteinase-13 (MMP-13), aggrecan, collagen Ⅱ, IL-1β, IL-6, tumour necrosis factor-alpha (TNF-α), and nuclear factor kappa-B (NF-κB) mRNA and protein expression in rat articular cartilage were examined by real-time quantitative polymerase chain reaction (RT-qPCR) and western blotting, respectively.

Results. Intra-articular injections of PRP significantly improved the structural integrity of the articular cartilage and enhanced the synthesis of glycosaminoglycans. PRP reduced MMP-13 protein level but increased aggrecan and collagen Ⅱ protein levels in articular cartilage of OA rats. OA-induced increase in serum IL-1β, IL-6, and TNF-α concentrations as well as increased MMP-13, and decreased collagen II mRNA levels were reversed by the administration of PRP. OA increased IL-1β, TNF-α,  and NF-κB mRNA expression in rat articular cartilage whereas PRP administration ameliorated these changes. Moreover, in the articular tissue of OA-induced rats the increased OPG protein level was further elevated by PRP injections whereas the protein level of RANK did not change. The increase in the protein level of RANKL in OA-induced articular tissue was offset by PRP administration. PRP elevated OPG mRNA expression and the OPG/RANKL mRNA ratio, but reduced RANKL mRNA expression and the RANKL/RANK mRNA ratio in the articular tissue of OA-induced rats.

Conclusions. PRP mitigates cartilage degradation and inflammation in experimental knee OA by regulating the OPG/RANKL/RANK signalling system.

Article available in PDF format

View PDF Download PDF file

References

  1. Abramoff B, Caldera FE. Osteoarthritis: pathology, diagnosis, and treatment options. Med Clin North Am. 2020; 104(2): 293–311.
  2. Xia B, Zhang J, Hu S, et al. Osteoarthritis pathogenesis: a review of molecular mechanisms. Calcif Tissue Int. 2014; 95(6): 495–505.
  3. Peng Z, Sun H, Bunpetch V, et al. The regulation of cartilage extracellular matrix homeostasis in joint cartilage degeneration and regeneration. Biomaterials. 2021; 268: 120555.
  4. Wang J, Verdonk P, Elewaut D, et al. Homeostasis of the extracellular matrix of normal and osteoarthritic human articular cartilage chondrocytes in vitro. Osteoarthritis Cartilage. 2003; 11(11): 801–809.
  5. Li X, Jia F, Zhu Z, et al. Lixisenatide attenuates advanced glycation end products (AGEs)-induced degradation of extracellular matrix in human primary chondrocytes. Artif Cells Nanomed Biotechnol. 2019; 47(1): 1256–1264.
  6. Zhang P, Chen Y, Zhao H, et al. Protective effects of alogliptin against TNF-α-induced degradation of extracellular matrix in human chondrocytes. Int Immunopharmacol. 2019; 68: 179–184.
  7. Taruc-Uy RL, Lynch SA. Diagnosis and treatment of osteoarthritis. Prim Care. 2013; 40(4): 821–36, vii.
  8. Alves R, Grimalt R. A review of platelet-rich plasma: history, biology, mechanism of action, and classification. Skin Appendage Disord. 2018; 4(1): 18–24.
  9. Gupta S, Paliczak A, Delgado D. Evidence-based indications of platelet-rich plasma therapy. Expert Rev Hematol. 2021; 14(1): 97–108.
  10. Bos-Mikich A, de Oliveira R, Frantz N. Platelet-rich plasma therapy and reproductive medicine. J Assist Reprod Genet. 2018; 35(5): 753–756.
  11. Nurden AT. Platelets, inflammation and tissue regeneration. Thromb Haemost. 2011; 105 Suppl 1: S13–S33.
  12. Palma-Barqueros V, Revilla N, Sánchez A, et al. Inherited platelet disorders: an updated overview. Int J Mol Sci. 2021; 22(9).
  13. Bennell KL, Hunter DJ, Paterson KL. Platelet-rich plasma for the management of hip and knee osteoarthritis. Curr Rheumatol Rep. 2017; 19(5): 24.
  14. Chen Pu, Huang L, Ma Y, et al. Intra-articular platelet-rich plasma injection for knee osteoarthritis: a summary of meta-analyses. J Orthop Surg Res. 2019; 14(1): 385.
  15. Sakata R, Reddi AH. Platelet-Rich Plasma Modulates Actions on Articular Cartilage Lubrication and Regeneration. Tissue Eng Part B Rev. 2016; 22(5): 408–419.
  16. Hooshiar SH, Tobeiha M, Jafarnejad S. Soy isoflavones and bone health: focus on the RANKL/RANK/OPG pathway. Biomed Res Int. 2022; 2022: 8862278.
  17. Miyamoto Y, Hasegawa T, Hongo H, et al. Histochemical assessment of osteoclast-like giant cells in Rankl(-/-) mice. J Oral Biosci. 2023; 65(2): 175–185.
  18. Ono T, Hayashi M, Sasaki F, et al. RANKL biology: bone metabolism, the immune system, and beyond. Inflamm Regen. 2020; 40: 2.
  19. Udagawa N, Koide M, Nakamura M, et al. Osteoclast differentiation by RANKL and OPG signaling pathways. J Bone Miner Metab. 2021; 39(1): 19–26.
  20. Yu T, Liu X, Jiang M, et al. Cucumber seed polypeptides regulate RANKL-induced osteoclastogenesis through OPG/RANKL/RANK and NF-κB. In Vitro Cell Dev Biol Anim. 2024; 60(1): 54–66.
  21. Kwan Tat S, Amiable N, Pelletier JP, et al. Modulation of OPG, RANK and RANKL by human chondrocytes and their implication during osteoarthritis. Rheumatology (Oxford). 2009; 48(12): 1482–1490.
  22. Wu W, Song K, Chen G, et al. Ganoderic acid A improves osteoarthritis by regulating RANKL/OPG ratio. Chem Biol Drug Des. 2022; 100(3): 313–319.
  23. Asjid R, Faisal T, Qamar K, et al. Effect of platelet-rich plasma on mankin scoring in chemically-induced animal model of osteoarthritis. J Coll Physicians Surg Pak. 2019; 29(11): 1067–1071.
  24. Kim JL, Moon CW, Son YS, et al. Combined Effect of Bilateral Ovariectomy and Anterior Cruciate Ligament Transection With Medial Meniscectomy on the Development of Osteoarthritis Model. Ann Rehabil Med. 2016; 40(4): 583–591.
  25. Wang F, Liu J, Chen X, et al. IL-1β receptor antagonist (IL-1Ra) combined with autophagy inducer (TAT-Beclin1) is an effective alternative for attenuating extracellular matrix degradation in rat and human osteoarthritis chondrocytes. Arthritis Res Ther. 2019; 21(1): 171.
  26. Khader A, Alquran H. Automated prediction of osteoarthritis level in human osteochondral tissue using histopathological images. Bioengineering (Basel). 2023; 10(7).
  27. Tschopp M, Pfirrmann CWA, Brunner F, et al. A randomized trial of intra-articular injection therapy for knee osteoarthritis. Invest Radiol. 2023; 58(5): 355–362.
  28. Mathiessen A, Conaghan PG. Synovitis in osteoarthritis: current understanding with therapeutic implications. Arthritis Res Ther. 2017; 19(1): 18.
  29. Hu H, Yang B, Li Y, et al. Blocking of the P2X7 receptor inhibits the activation of the MMP-13 and NF-κB pathways in the cartilage tissue of rats with osteoarthritis. Int J Mol Med. 2016; 38(6): 1922–1932.
  30. van Spil WE, Szilagyi IA. Osteoarthritis year in review 2019: biomarkers (biochemical markers). Osteoarthritis Cartilage. 2020; 28(3): 296–315.
  31. Choi MC, Jo J, Park J, et al. NF-κB signaling pathways in osteoarthritic cartilage destruction. Cells. 2019; 8(7).
  32. Zhu S, Gu Ye, Wang W, et al. Sitagliptin ameliorates advanced glycation end-product (AGE)-induced degradation of extracellular matrix in human primary chondrocytes. Am J Transl Res. 2019; 11(5): 2775–2783.
  33. Goldring MB, Otero M. Inflammation in osteoarthritis. Curr Opin Rheumatol. 2011; 23(5): 471–478.
  34. Zhao H, Zhu W, Mao W, et al. Platelet-rich plasma inhibits Adriamycin-induced inflammation via blocking the NF-κB pathway in articular chondrocytes. Mol Med. 2021; 27(1): 66.
  35. Zhuo F, Jia X, Wang Z, et al. Platelet-rich plasma alleviates knee arthritis in rats by inhibiting p65. Cell Tissue Bank. 2024; 25(2): 463–473.
  36. Yin WJ, Xu HT, Sheng JG, et al. Advantages of pure platelet-rich plasma compared with leukocyte- and platelet-rich plasma in treating rabbit knee osteoarthritis. Med Sci Monit. 2016; 22: 1280–1290.
  37. Poole AR, Kobayashi M, Yasuda T, et al. Type II collagen degradation and its regulation in articular cartilage in osteoarthritis. Ann Rheum Dis. 2002; 61 Suppl 2(Suppl 2): ii78–ii81.
  38. Ouyang Z, Dong L, Yao F, et al. Cartilage-related collagens in osteoarthritis and rheumatoid arthritis: from pathogenesis to therapeutics. Int J Mol Sci. 2023; 24(12).
  39. Jansen IDC, Hollander AP, Buttle DJ, et al. Type II and VI collagen in nasal and articular cartilage and the effect of IL-1alpha on the distribution of these collagens. J Mol Histol. 2010; 41(1): 9–17.
  40. Huang C, Liao G, Han J, et al. Edaravone suppresses degradation of type II collagen. Biochem Biophys Res Commun. 2016; 473(4): 840–844.
  41. Tiku ML, Madhan B. Preserving the longevity of long-lived type II collagen and its implication for cartilage therapeutics. Ageing Res Rev. 2016; 28: 62–71.
  42. Mehana ESE, Khafaga AF, El-Blehi SS. The role of matrix metalloproteinases in osteoarthritis pathogenesis: an updated review. Life Sci. 2019; 234: 116786.
  43. Yoshihara Y, Nakamura H, Obata K, et al. Matrix metalloproteinases and tissue inhibitors of metalloproteinases in synovial fluids from patients with rheumatoid arthritis or osteoarthritis. Ann Rheum Dis. 2000; 59(6): 455–461.
  44. Hu N, Gong X, Yin S, et al. Saxagliptin suppresses degradation of type II collagen and aggrecan in primary human chondrocytes: a therapeutic implication in osteoarthritis. Artif Cells Nanomed Biotechnol. 2019; 47(1): 3239–3245.
  45. Hu Q, Ecker M. Overview of MMP-13 as a promising target for the treatment of osteoarthritis. Int J Mol Sci. 2021; 22(4).
  46. Wang X, Fan J, Ding X, et al. Tanshinone i inhibits IL-1β-induced apoptosis, inflammation and extracellular matrix degradation in chondrocytes CHON-001 cells and attenuates murine osteoarthritis. Drug Des Devel Ther. 2019; 13: 3559–3568.
  47. Zhang Y, Lin J, Zhou X, et al. Melatonin prevents osteoarthritis-induced cartilage degradation via targeting microrna-140. Oxid Med Cell Longev. 2019; 2019: 9705929.
  48. Yin W, Xu H, Sheng J, et al. Comparative evaluation of the effects of platelet‑rich plasma formulations on extracellular matrix formation and the NF‑κB signaling pathway in human articular chondrocytes. Mol Med Rep. 2017; 15(5): 2940–2948.
  49. van Buul GM, Koevoet WLM, Kops N, et al. Platelet-rich plasma releasate inhibits inflammatory processes in osteoarthritic chondrocytes. Am J Sports Med. 2011; 39(11): 2362–2370.
  50. Du J, Wang Y, Wu C, et al. Targeting bone homeostasis regulation: potential of traditional Chinese medicine flavonoids in the treatment of osteoporosis. Front Pharmacol. 2024; 15: 1361864.
  51. Ma L, Liu X, Sun G. Aspirin reduces the incidence of postmenopausal osteoporosis in rats through OPG-RANKL-RANK signaling pathway. Minerva Endocrinol. 2019; 44(4): 408–410.
  52. Ritchlin CT, Schwarz EM, O'Keefe RJ, et al. RANK, RANKL and OPG in inflammatory arthritis and periprosthetic osteolysis. J Musculoskelet Neuronal Interact. 2004; 4(3): 276–284.
  53. Upton AR, Holding CA, Dharmapatni AA, et al. The expression of RANKL and OPG in the various grades of osteoarthritic cartilage. Rheumatol Int. 2012; 32(2): 535–540.
  54. Kovács B, Vajda E, Nagy EE. Regulatory effects and interactions of the wnt and OPG-RANKL-RANK signaling at the bone-cartilage interface in osteoarthritis. Int J Mol Sci. 2019; 20(18).
  55. Bolon B, Grisanti M, Villasenor K, et al. Generalized degenerative joint disease in osteoprotegerin (Opg) null mutant mice. Vet Pathol. 2015; 52(5): 873–882.
  56. Bucay N, Sarosi I, Dunstan CR, et al. osteoprotegerin-deficient mice develop early onset osteoporosis and arterial calcification. Genes Dev. 1998; 12(9): 1260–1268.
  57. Kong YY, Yoshida H, Sarosi I, et al. OPGL is a key regulator of osteoclastogenesis, lymphocyte development and lymph-node organogenesis. Nature. 1999; 397(6717): 315–323.
  58. Gao X, Wu Q, Zhang X, et al. Salvianolate ameliorates osteopenia and improves bone quality in prednisone-treated rheumatoid arthritis rats by regulating RANKL/RANK/OPG signaling. Front Pharmacol. 2021; 12: 710169.
  59. Liu WW, Xu ZM, Li ZQ, et al. RANKL, OPG and CTR mRNA expression in the temporomandibular joint in rheumatoid arthritis. Exp Ther Med. 2015; 10(3): 895–900.
  60. Kong YY, Feige U, Sarosi I, et al. Activated T cells regulate bone loss and joint destruction in adjuvant arthritis through osteoprotegerin ligand. Nature. 1999; 402(6759): 304–309.
  61. Zeng JZ, Wang ZZ, Ma LF, et al. Increased receptor activator of nuclear factor κβ ligand/osteoprotegerin ratio exacerbates cartilage destruction in osteoarthritis. Exp Ther Med. 2016; 12(4): 2778–2782.
  62. Bellido M, Lugo L, Roman-Blas JA, et al. Subchondral bone microstructural damage by increased remodelling aggravates experimental osteoarthritis preceded by osteoporosis. Arthritis Res Ther. 2010; 12(4): R152–1236.
  63. Sakao K, Takahashi KA, Mazda O, et al. Enhanced expression of interleukin-6, matrix metalloproteinase-13, and receptor activator of NF-kappaB ligand in cells derived from osteoarthritic subchondral bone. J Orthop Sci. 2008; 13(3): 202–210.