Advantages of Pure Platelet-Rich Plasma Compared with Leukocyte- and Platelet-Rich Plasma in Treating Rabbit Knee Osteoarthritis

Wen-Jing Yin, Hai-Tao Xu, Jia-Gen Sheng, Zhi-Quan An, Shang-Chun Guo, Xue-Tao Xie, Chang-Qing Zhang, Wen-Jing Yin, Hai-Tao Xu, Jia-Gen Sheng, Zhi-Quan An, Shang-Chun Guo, Xue-Tao Xie, Chang-Qing Zhang

Abstract

BACKGROUND Concentrated leukocytes in leukocyte- and platelet-rich plasma (L-PRP) may deliver increased levels of pro-inflammatory cytokines to activate the NF-κB signaling pathway, to counter the beneficial effects of growth factors on osteoarthritic cartilage. However, to date no relevant studies have substantiated that in vivo. MATERIAL AND METHODS Autologous L-PRP and pure platelet-rich plasma (P-PRP) were prepared, measured for componential composition, and injected intra-articularly after 4, 5, and 6 weeks post-anterior cruciate ligament transection. Caffeic acid phenethyl ester (CAPE) was injected intraperitoneally to inhibit NF-κB activation. All rabbits were sacrificed after 8 weeks postoperative. Enzyme-linked immunosorbent assays were performed to determine interleukin 1β (IL-1β) and prostaglandin E2 (PGE2) concentrations in the synovial fluid, Indian ink staining was performed for gross morphological assessment, and hematoxylin and eosin staining and toluidine blue staining were performed for histological assessment. RESULTS Compared with L-PRP, P-PRP injections achieved better outcomes regarding the prevention of cartilage destruction, preservation of cartilaginous matrix, and reduction of IL-1β and PGE2 concentrations. CAPE injections reversed the increased IL-1β and PGE2 concentrations in the synovial fluid after L-PRP injections and improved the outcome of L-PRP injections to a level similar to P-PRP injections, while they had no influence on the therapeutic efficacy of P-PRP injections. CONCLUSIONS Concentrated leukocytes in L-PRP may release increased levels of pro-inflammatory cytokines to activate the NF-κB signaling pathway, to counter the beneficial effects of growth factors on osteoarthritic cartilage, and finally, result in a inferior efficacy of L-PRP to P-PRP for the treatment of osteoarthritis.

Figures

Figure 1
Figure 1
Study design. L-PRP – leukocyte- and platelet-rich plasma; P-PRP – pure platelet-rich plasma; CAPE – caffeic acid phenethyl ester.
Figure 2
Figure 2
Leukocyte and platelet concentrations in platelet-rich plasma formulations used in the study. (A) At each time point, the leukocyte concentrations in leukocyte- and platelet-rich plasma (L-PRP) used in the L-PRP group and L-PRP+ caffeic acid phenethyl ester (CAPE) group were higher than in pure platelet-rich plasma (P-PRP), which was used in the P-PRP group and P-PRP+CAPE group. (B) L-PRP and P-PRP were similar in platelet concentrations. Boxes and error bars represent mean ± standard deviation (n=10); * p<0.05 compared with L-PRP used in the L-PRP group at the same time point; #p<0.05 compared with L-PRP used in the L-PRP+CAPE group at the same time point.
Figure 3
Figure 3
Cytokine concentrations in platelet-rich plasma formulations used in the study. (A, B) At each time point, the concentrations of interleukin-1β (IL-1β); (A) Tumor necrosis factor-α (TNF-α). (B) Leukocyte- and platelet-rich plasma (L-PRP) used in the L-PRP group and L-PRP+ caffeic acid phenethyl ester (CAPE) group were higher than in pure platelet-rich plasma (P-PRP), which was used in the P-PRP group and P-PRP+CAPE group; (C, D) L-PRP and P-PRP were similar in the concentrations of platelet-derived growth factor AB (PDGF-AB). (C) Transforming growth factor β1 (TGF-β1). (D) Boxes and error bars represent mean ± standard deviation (n=10); * p<0.05 compared with L-PRP used in the L-PRP group at the same time point; #p<0.05 compared with L-PRP used in the L-PRP+CAPE group at the same time point.
Figure 4
Figure 4
Correlations between components of platelet-rich plasma formulations. There was a significantly positive correlation between leukocyte concentration and IL-1β concentration (A) and TNF-α (B) concentration, and between platelet concentration and PDGF-AB concentration (C) and TGF-β1 concentration (D). The correlations between leukocyte concentration and PDGF-AB (E) and TGF-β1 (F) concentrations and the correlations between platelet concentration and IL-1β (G) and TNF-α (H) concentrations were not significant. IL-1β, interleukin-1β; TNF-α, tumor necrosis factor-α; PDGF-AB, platelet-derived growth factor AB; TGF-β1, transforming growth factor-β1.
Figure 5
Figure 5
Gross morphological assessment of cartilage degeneration. Gross morphological assessment was performed on both the medial and lateral sides of femoral condyles according to the following criteria after Indian ink staining: grade 1, no staining by Indian ink; grade 2, surface retains ink as elongated specks or light gray patches; grade 3, surface retains ink as intense black patches; grade 4, loss of cartilage exposing the sub-cartilaginous bone; grade 4a, 0 mm p<0.05 compared with the control group; #p<0.05 compared with the L-PRP group (n=40).
Figure 6
Figure 6
Interleukin 1β (IL-1β) and prostaglandin E2 (PGE2) concentrations in the synovial fluid. IL-1β (A) and PGE2 (B) concentrations in the synovial fluid were quantified by enzyme-linked immunosorbent assay. The mean IL-1β and PGE2 concentrations in the synovial fluid collected from rabbits of the L-PRP group were significantly higher than the control group, which, in turn, was significantly higher than the P-PRP group, L-PRP+CAPE group, and P-PRP+CAPE group, which were similar compared with each other. L-PRP, leukocyte- and platelet-rich plasma; P-PRP, pure platelet-rich plasma; CAPE, caffeic acid phenethyl ester. Boxes and error bars represent mean ± standard deviation (n=20); * p<0.05 compared with the control group; #p<0.05 compared with the L-PRP group.
Figure 7
Figure 7
Hematoxylin and eosin staining and toluidine blue staining for histological assessment. Representative HE stained sections (A, C, E, G, I) and toluidine blue stained sections (B, D, F, H, J) of femoral condyles after 8 weeks postoperative. L-PRP – leukocyte- and platelet-rich plasma; P-PRP – pure platelet-rich plasma; CAPE – caffeic acid phenethyl ester. Scales represent 200 μm.

References

    1. Woolf AD, Pfleger B. Burden of major musculoskeletal conditions. Bull World Health Organ. 2003;81:646–56.
    1. Santaguida PL, Hawker GA, Hudak PL, et al. Patient characteristics affecting the prognosis of total hip and knee joint arthroplasty: A systematic review. Can J Surg. 2008;51:428–36.
    1. Muir H. The chondrocyte, architect of cartilage. Biomechanics, structure, function and molecular biology of cartilage matrix macromolecules. Bioessays. 1995;17:1039–48.
    1. Chevalier X, Eymard F, Richette P. Biologic agents in osteoarthritis: Hopes and disappointments. Nat Rev Rheumatol. 2013;9:400–10.
    1. Castillo TN, Pouliot MA, Kim HJ, Dragoo JL. Comparison of growth factor and platelet concentration from commercial platelet-rich plasma separation systems. Am J Sports Med. 2011;39:266–71.
    1. Magalon J, Bausset O, Serratrice N, et al. Characterization and comparison of 5 platelet-rich plasma preparations in a single-donor model. Arthroscopy. 2014;30:629–38.
    1. Appel TR, Potzsch B, Muller J, et al. Comparison of three different preparations of platelet concentrates for growth factor enrichment. Clin Oral Implants Res. 2002;13:522–28.
    1. Ellman MB, An HS, Muddasani P, Im HJ. Biological impact of the fibroblast growth factor family on articular cartilage and intervertebral disc homeostasis. Gene. 2008;420:82–89.
    1. Brandl A, Angele P, Roll C, et al. Influence of the growth factors PDGF-BB, TGF-beta1 and bFGF on the replicative aging of human articular chondrocytes during in vitro expansion. J Orthop Res. 2010;28:354–60.
    1. Fortier LA, Barker JU, Strauss EJ, et al. The role of growth factors in cartilage repair. Clin Orthop Relat Res. 2011;469:2706–15.
    1. Kon E, Mandelbaum B, Buda R, et al. Platelet-rich plasma intra-articular injection versus hyaluronic acid viscosupplementation as treatments for cartilage pathology: From early degeneration to osteoarthritis. Arthroscopy. 2011;27:1490–501.
    1. Filardo G, Kon E, Buda R, et al. Platelet-rich plasma intra-articular knee injections for the treatment of degenerative cartilage lesions and osteoarthritis. Knee Surg Sports Traumatol Arthrosc. 2011;19:528–35.
    1. Kon E, Buda R, Filardo G, et al. Platelet-rich plasma: intra-articular knee injections produced favorable results on degenerative cartilage lesions. Knee Surg Sports Traumatol Arthrosc. 2010;18:472–79.
    1. Dohan Ehrenfest DM, Rasmusson L, Albrektsson T. Classification of platelet concentrates: from pure platelet-rich plasma (P-PRP) to leucocyte- and platelet-rich fibrin (L-PRF) Trends Biotechnol. 2009;27:158–67.
    1. Sundman EA, Cole BJ, Fortier LA. Growth factor and catabolic cytokine concentrations are influenced by the cellular composition of platelet-rich plasma. Am J Sports Med. 2011;39:2135–40.
    1. Burguera EF, Vela-Anero A, Magalhaes J, et al. Effect of hydrogen sulfide sources on inflammation and catabolic markers on interleukin 1beta-stimulated human articular chondrocytes. Osteoarthritis Cartilage. 2014;22:1026–35.
    1. Campbell KA, Minashima T, Zhang Y, et al. Annexin A6 interacts with p65 and stimulates NF-kappaB activity and catabolic events in articular chondrocytes. Arthritis Rheum. 2013;65:3120–29.
    1. Davidson RK, Jupp O, de Ferrars R, et al. Sulforaphane represses matrix-degrading proteases and protects cartilage from destruction in vitro and in vivo. Arthritis Rheum. 2013;65:3130–40.
    1. Cavallo C, Filardo G, Mariani E, et al. Comparison of platelet-rich plasma formulations for cartilage healing: An in vitro study. J Bone Joint Surg Am. 2014;96:423–29.
    1. Yoshioka M, Coutts RD, Amiel D, Hacker SA. Characterization of a model of osteoarthritis in the rabbit knee. Osteoarthritis Cartilage. 1996;4:87–98.
    1. Sanchez M, Anitua E, Azofra J, et al. Intra-articular injection of an autologous preparation rich in growth factors for the treatment of knee OA: A retrospective cohort study. Clin Exp Rheumatol. 2008;26:910–13.
    1. Napolitano M, Matera S, Bossio M, et al. Autologous platelet gel for tissue regeneration in degenerative disorders of the knee. Blood Transfus. 2012;10:72–77.
    1. Spakova T, Rosocha J, Lacko M, et al. Treatment of knee joint osteoarthritis with autologous platelet-rich plasma in comparison with hyaluronic acid. Am J Phys Med Rehabil. 2012;91:411–17.
    1. Sanchez M, Fiz N, Azofra J, et al. A randomized clinical trial evaluating plasma rich in growth factors (PRGF-Endoret) versus hyaluronic acid in the short-term treatment of symptomatic knee osteoarthritis. Arthroscopy. 2012;28:1070–78.
    1. Altug ME, Serarslan Y, Bal R, et al. Caffeic acid phenethyl ester protects rabbit brains against permanent focal ischemia by antioxidant action: A biochemical and planimetric study. Brain Res. 2008;1201:135–42.
    1. Jia WT, Zhang CQ, Wang JQ, et al. The prophylactic effects of platelet-leucocyte gel in osteomyelitis: An experimental study in a rabbit model. J Bone Joint Surg Br. 2010;92:304–10.
    1. Amiel D, Toyoguchi T, Kobayashi K, et al. Long-term effect of sodium hyaluronate (Hyalgan) on osteoarthritis progression in a rabbit model. Osteoarthritis Cartilage. 2003;11:636–43.
    1. Batten ML, Hansen JC, Dahners LE. Influence of dosage and timing of application of platelet-derived growth factor on early healing of the rat medial collateral ligament. J Orthop Res. 1996;14:736–41.
    1. Torricelli P, Fini M, Filardo G, et al. Regenerative medicine for the treatment of musculoskeletal overuse injuries in competition horses. Int Orthop. 2011;35:1569–76.
    1. Weibrich G, Hansen T, Kleis W, et al. Effect of platelet concentration in platelet-rich plasma on peri-implant bone regeneration. Bone. 2004;34:665–71.
    1. Anitua E, Sanchez M, Zalduendo MM, et al. Fibroblastic response to treatment with different preparations rich in growth factors. Cell Prolif. 2009;42:162–70.
    1. Heldin CH, Eriksson U, Ostman A. New members of the platelet-derived growth factor family of mitogens. Arch Biochem Biophys. 2002;398:284–90.
    1. Yaeger PC, Masi TL, de Ortiz JL, et al. Synergistic action of transforming growth factor-beta and insulin-like growth factor-I induces expression of type II collagen and aggrecan genes in adult human articular chondrocytes. Exp Cell Res. 1997;237:318–25.
    1. Stewart K, Pabbruwe M, Dickinson S, et al. The effect of growth factor treatment on meniscal chondrocyte proliferation and differentiation on polyglycolic acid scaffolds. Tissue Eng. 2007;13:271–80.
    1. Redini F, Mauviel A, Pronost S, et al. Transforming growth factor beta exerts opposite effects from interleukin-1 beta on cultured rabbit articular chondrocytes through reduction of interleukin-1 receptor expression. Arthritis Rheum. 1993;36:44–50.
    1. Berg U, Gustafsson T, Sundberg CJ, et al. Local changes in the insulin-like growth factor system in human skeletal muscle assessed by microdialysis and arterio-venous differences technique. Growth Horm IGF Res. 2006;16:217–23.
    1. Novak ML, Koh TJ. Macrophage phenotypes during tissue repair. J Leukoc Biol. 2013;93:875–81.
    1. Li H, Hicks JJ, Wang L, et al. Customized platelet-rich plasma with transforming growth factor beta1 neutralization antibody to reduce fibrosis in skeletal muscle. Biomaterials. 2016;87:147–56.
    1. Bielecki T, Dohan Ehrenfest DM, et al. The role of leukocytes from L-PRP/L-PRF in wound healing and immune defense: New perspectives. Curr Pharm Biotechnol. 2012;13:1153–62.
    1. Murray PJ, Wynn TA. Protective and pathogenic functions of macrophage subsets. Nat Rev Immunol. 2011;11:723–37.
    1. Russell RP, Apostolakos J, Hirose T, et al. Variability of platelet-rich plasma preparations. Sports Med Arthrosc. 2013;21:186–90.
    1. McCarrel TM, Minas T, Fortier LA. Optimization of leukocyte concentration in platelet-rich plasma for the treatment of tendinopathy. J Bone Joint Surg Am. 2012;94:e143(1–8).
    1. Wood DD, Ihrie EJ, Dinarello CA, Cohen PL. Isolation of an interleukin-1-like factor from human joint effusions. Arthritis Rheum. 1983;26:975–83.
    1. Saklatvala J. Tumour necrosis factor alpha stimulates resorption and inhibits synthesis of proteoglycan in cartilage. Nature. 1986;322:547–49.
    1. Pujol JP, Chadjichristos C, Legendre F, et al. Interleukin-1 and transforming growth factor-beta 1 as crucial factors in osteoarthritic cartilage metabolism. Connect Tissue Res. 2008;49:293–97.
    1. Liu Y, Peng H, Meng Z, Wei M. Correlation of IL-17 level in synovia and severity of knee osteoarthritis. Med Sci Monit. 2015;21:1732–36.
    1. Henderson B, Pettipher ER. Arthritogenic actions of recombinant IL-1 and tumour necrosis factor alpha in the rabbit: Evidence for synergistic interactions between cytokines in vivo. Clin Exp Immunol. 1989;75:306–10.
    1. Bacconnier L, Jorgensen C, Fabre S. Erosive osteoarthritis of the hand: Clinical experience with anakinra. Ann Rheum Dis. 2009;68:1078–79.
    1. Grunke M, Schulze-Koops H. Successful treatment of inflammatory knee osteoarthritis with tumour necrosis factor blockade. Ann Rheum Dis. 2006;65:555–56.
    1. Ou Y, Tan C, An H, et al. Selective COX-2 inhibitor ameliorates osteoarthritis by repressing apoptosis of chondrocyte. Med Sci Monit. 2012;18(6):BR247–52.
    1. Pereira RC, Scaranari M, Benelli R, et al. Dual effect of platelet lysate on human articular cartilage: A maintenance of chondrogenic potential and a transient proinflammatory activity followed by an inflammation resolution. Tissue Eng Part A. 2013;19:1476–88.
    1. van Buul GM, Koevoet WL, Kops N, et al. Platelet-rich plasma releasate inhibits inflammatory processes in osteoarthritic chondrocytes. Am J Sports Med. 2011;39:2362–70.
    1. Ledoux AC, Perkins ND. NF-kappaB and the cell cycle. Biochem Soc Trans. 2014;42:76–81.
    1. Marcu KB, Otero M, Olivotto E, et al. NF-κB signaling: multiple angles to target OA. Curr Drug Targets. 2010;11:599–613.

Source: PubMed

3
订阅