DOI QR코드

DOI QR Code

Decay-Accelerating Factor Differentially Associates With Complement-Mediated Damage in Synovium After Meniscus Tear as Compared to Anterior Cruciate Ligament Injury

  • V. Michael Holers (Division of Rheumatology, School of Medicine, University of Colorado, Anschutz Medical Campus) ;
  • Rachel M. Frank (Department of Orthopedics and the Colorado Program for Musculoskeletal Research, School of Medicine, University of Colorado, Anschutz Medical Campus) ;
  • Michael Zuscik (Department of Orthopedics and the Colorado Program for Musculoskeletal Research, School of Medicine, University of Colorado, Anschutz Medical Campus) ;
  • Carson Keeter (Department of Orthopedics and the Colorado Program for Musculoskeletal Research, School of Medicine, University of Colorado, Anschutz Medical Campus) ;
  • Robert I. Scheinman (Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Anschutz Medical Campus) ;
  • Christopher Striebich (Division of Rheumatology, School of Medicine, University of Colorado, Anschutz Medical Campus) ;
  • Dmitri Simberg (Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Anschutz Medical Campus) ;
  • Michael R. Clay (Department of Pathology, School of Medicine, University of Colorado, Anschutz Medical Campus) ;
  • Larry W. Moreland (Division of Rheumatology, School of Medicine, University of Colorado, Anschutz Medical Campus) ;
  • Nirmal K. Banda (Division of Rheumatology, School of Medicine, University of Colorado, Anschutz Medical Campus)
  • 투고 : 2023.12.11
  • 심사 : 2024.04.08
  • 발행 : 2024.04.30

초록

We have reported that anterior cruciate ligament (ACL) injury leads to the differential dysregulation of the complement system in the synovium as compared to meniscus tear (MT) and proposed this as a mechanism for a greater post-injury prevalence of post traumatic osteoarthritis (PTOA). To explore additional roles of complement proteins and regulators, we determined the presence of decay-accelerating factor (DAF), C5b, and membrane attack complexes (MACs, C5b-9) in discarded surgical synovial tissue (DSST) collected during arthroscopic ACL reconstructive surgery, MT-related meniscectomy, osteoarthritis (OA)-related knee replacement surgery and normal controls. Multiplexed immunohistochemistry was used to detect and quantify complement proteins. To explore the involvement of body mass index (BMI), after these 2 injuries, we examined correlations among DAF, C5b, MAC and BMI. Using these approaches, we found that synovial cells after ACL injury expressed a significantly lower level of DAF as compared to MT (p<0.049). In contrast, C5b staining synovial cells were significantly higher after ACL injury (p<0.0009) and in OA DSST (p<0.039) compared to MT. Interestingly, there were significantly positive correlations between DAF & C5b (r=0.75, p<0.018) and DAF & C5b (r=0.64 p<0.022) after ACL injury and MT, respectively. The data support that DAF, which should normally dampen C5b deposition due to its regulatory activities on C3/C5 convertases, does not appear to exhibit that function in inflamed synovia following either ACL injury or MT. Ineffective DAF regulation may be an additional mechanism by which relatively uncontrolled complement activation damages tissue in these injury states.

키워드

과제정보

Authors are thankful to Mr. Andrew Clauw who was involved in routine tissue collection, processing, and preservation of DSST from ACL injury and MT. All authors are also thankful to Ms. Jennifer Seifert who catalogued ACL and MT related DSST samples and kept the patients record securely using RedCap. We are also thankful to Ms. Terrin Manes for providing histology sections from ACL and MT DSST for synovial membrane DAF, macrophage and fibroblast MIHC studies. Thanks to Ms. Elizabeth Smith in the AMC Histopathology Clinical core for helping to standardize DAF and immune cell-related antibodies along with their antigen retrieval procedures. We are also thankful to Dr. Kimberley Jordan, and Mr. Troy Schedin from the University of Colorado Human Immune Monitoring Shared Resource (HIMSR) group for MIHC related work and specifically for generating composite images and helping in analysis. Supported by the National Institutes of Health grant R01 AR51749 to VMH (PI) and NKB (Co-I) and by the Institutional Joint Biology Program pilot grant to NKB (PI) and RMF (Co-I).

참고문헌

  1. Brown TD, Johnston RC, Saltzman CL, Marsh JL, Buckwalter JA. Posttraumatic osteoarthritis: a first estimate of incidence, prevalence, and burden of disease. J Orthop Trauma 2006;20:739-744.
  2. Hsia AW, Jbeily EH, Mendez ME, Cunningham HC, Biris KK, Bang H, Lee CA, Loots GG, Christiansen BA. Post-traumatic osteoarthritis progression is diminished by early mechanical unloading and anti-inflammatory treatment in mice. Osteoarthritis Cartilage 2021;29:1709-1719.
  3. Dare D, Rodeo S. Mechanisms of post-traumatic osteoarthritis after ACL injury. Curr Rheumatol Rep 2014;16:448.
  4. Lohmander LS, Englund PM, Dahl LL, Roos EM. The long-term consequence of anterior cruciate ligament and meniscus injuries: osteoarthritis. Am J Sports Med 2007;35:1756-1769.
  5. Racine J, Aaron RK. Post-traumatic osteoarthritis after ACL injury. R I Med J (2013) 2014;97:25-28.
  6. Wang LJ, Zeng N, Yan ZP, Li JT, Ni GX. Post-traumatic osteoarthritis following ACL injury. Arthritis Res Ther 2020;22:57.
  7. Ratzlaff CR, Liang MH. New developments in osteoarthritis. Prevention of injury-related knee osteoarthritis: opportunities for the primary and secondary prevention of knee osteoarthritis. Arthritis Res Ther 2010;12:215.
  8. Holers VM, Frank RM, Clauw A, Seifert J, Zuscik M, Asokan S, Striebich C, Clay MR, Moreland LW, Banda NK. Potential causal role of synovial complement system activation in the development of post-traumatic osteoarthritis after anterior cruciate ligament injury or meniscus tear. Front Immunol 2023;14:1146563.
  9. Toomey CB, Cauvi DM, Pollard KM. The role of decay accelerating factor in environmentally induced and idiopathic systemic autoimmune disease. Autoimmune Dis 2014;2014:452853.
  10. Frank MM, Sullivan KE. Chapter 38 - Deficiencies of the complement system. In: Stiehm's Immune Deficiencies. Sullivan KE, Stiehm ER, eds. Amsterdam: Academic Press; 2014. p.731-763. 
  11. Medof ME, Walter EI, Rutgers JL, Knowles DM, Nussenzweig V. Identification of the complement decay-accelerating factor (DAF) on epithelium and glandular cells and in body fluids. J Exp Med 1987;165:848-864.
  12. Lukacik P, Roversi P, White J, Esser D, Smith GP, Billington J, Williams PA, Rudd PM, Wormald MR, Harvey DJ, et al. Complement regulation at the molecular level: the structure of decay-accelerating factor. Proc Natl Acad Sci USA 2004;101:1279-1284.
  13. Hamann J, Vogel B, van Schijndel GM, van Lier RA. The seven-span transmembrane receptor CD97 has a cellular ligand (CD55, DAF). J Exp Med 1996;184:1185-1189.
  14. Karpus ON, Heutinck KM, Wijnker PJ, Tak PP, Hamann J. Triggering of the dsRNA sensors TLR3, MDA5, and RIG-I induces CD55 expression in synovial fibroblasts. PLoS One 2012;7:e35606.
  15. Nicholson-Weller A, Burge J, Austen KF. Purification from guinea pig erythrocyte stroma of a decay-accelerating factor for the classical c3 convertase, C4b,2a. J Immunol 1981;127:2035-2039.
  16. Pangburn MK, Schreiber RD, Muller-Eberhard HJ. Deficiency of an erythrocyte membrane protein with complement regulatory activity in paroxysmal nocturnal hemoglobinuria. Proc Natl Acad Sci U S A 1983;80:5430-5434.
  17. Medof ME, Kinoshita T, Nussenzweig V. Inhibition of complement activation on the surface of cells after incorporation of decay-accelerating factor (DAF) into their membranes. J Exp Med 1984;160:1558-1578.
  18. Lublin DM, Atkinson JP. Decay-accelerating factor: biochemistry, molecular biology, and function. Annu Rev Immunol 1989;7:35-58.
  19. Geller A, Yan J. The role of membrane bound complement regulatory proteins in tumor development and cancer immunotherapy. Front Immunol 2019;10:1074.
  20. Dho SH, Lim JC, Kim LK. Beyond the role of CD55 as a complement component. Immune Netw 2018;18:e11.
  21. Naraoka T, Ishibashi Y, Tsuda E, Yamamoto Y, Kusumi T, Kakizaki I, Toh S. Time-dependent gene expression and immunohistochemical analysis of the injured anterior cruciate ligament. Bone Joint Res 2012;1:238-244.
  22. Karpus ON, Kiener HP, Niederreiter B, Yilmaz-Elis AS, van der Kaa J, Ramaglia V, Arens R, Smolen JS, Botto M, Tak PP, et al. CD55 deposited on synovial collagen fibers protects from immune complex-mediated arthritis. Arthritis Res Ther 2015;17:6.
  23. Tarkowski A, Trollmo C, Seifert PS, Hansson GK. Expression of decay-accelerating factor on synovial lining cells in inflammatory and degenerative arthritides. Rheumatol Int 1992;12:201-205.
  24. Krenn V, Perino G, Ruther W, Krenn VT, Huber M, Hugle T, Najm A, Muller S, Boettner F, Pessler F, et al. 15 Years of the histopathological synovitis score, further development and review: a diagnostic score for rheumatology and orthopaedics. Pathol Res Pract 2017;213:874-881.
  25. Banda NK, Deane KD, Bemis EA, Strickland C, Seifert J, Jordan K, Goldman K, Morgan BP, Moreland LW, Lewis MJ, et al. Analysis of complement gene expression, clinical associations, and biodistribution of complement proteins in the synovium of early rheumatoid arthritis patients reveals unique pathophysiologic features. J Immunol 2022;208:2482-2496.
  26. Nasu J, Mizuno M, Uesu T, Takeuchi K, Inaba T, Ohya S, Kawada M, Shimo K, Okada H, Fujita T, et al. Cytokine-stimulated release of decay-accelerating factor (DAF;CD55) from HT-29 human intestinal epithelial cells. Clin Exp Immunol 1998;113:379-385.
  27. Cocuzzi ET, Bardenstein DS, Stavitsky A, Sundarraj N, Medof ME. Upregulation of DAF (CD55) on orbital fibroblasts by cytokines. Differential effects of TNF-beta and TNF-alpha. Curr Eye Res 2001;23:86-92.
  28. Cuellar VG, Cuellar JM, Golish SR, Yeomans DC, Scuderi GJ. Cytokine profiling in acute anterior cruciate ligament injury. Arthroscopy 2010;26:1296-1301.
  29. Bigoni M, Turati M, Sacerdote P, Gaddi D, Piatti M, Castelnuovo A, Franchi S, Gandolla M, Pedrocchi A, Omeljaniuk RJ, et al. Characterization of synovial fluid cytokine profiles in chronic meniscal tear of the knee. J Orthop Res 2017;35:340-346.
  30. Spiller OB, Criado-Garcia O, Rodriguez De Cordoba S, Morgan BP. Cytokine-mediated up-regulation of CD55 and CD59 protects human hepatoma cells from complement attack. Clin Exp Immunol 2000;121:234-241.
  31. Martel-Pelletier J, McCollum R, DiBattista J, Faure MP, Chin JA, Fournier S, Sarfati M, Pelletier JP. The interleukin-1 receptor in normal and osteoarthritic human articular chondrocytes. Identification as the type I receptor and analysis of binding kinetics and biologic function. Arthritis Rheum 1992;35:530-540.
  32. Sadouk MB, Pelletier JP, Tardif G, Kiansa K, Cloutier JM, Martel-Pelletier J. Human synovial fibroblasts coexpress IL-1 receptor type I and type II mRNA. The increased level of the IL-1 receptor in osteoarthritic cells is related to an increased level of the type I receptor. Lab Invest 1995;73:347-355.
  33. Sellam J, Berenbaum F. The role of synovitis in pathophysiology and clinical symptoms of osteoarthritis. Nat Rev Rheumatol 2010;6:625-635.
  34. Loeser RF, Goldring SR, Scanzello CR, Goldring MB. Osteoarthritis: a disease of the joint as an organ. Arthritis Rheum 2012;64:1697-1707.
  35. 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;75:306-310.
  36. Berger M, Medof ME. Increased expression of complement decay-accelerating factor during activation of human neutrophils. J Clin Invest 1987;79:214-220.
  37. Wang Q, Rozelle AL, Lepus CM, Scanzello CR, Song JJ, Larsen DM, Crish JF, Bebek G, Ritter SY, Lindstrom TM, et al. Identification of a central role for complement in osteoarthritis. Nat Med 2011;17:1674-1679.
  38. Liu J, Miwa T, Hilliard B, Chen Y, Lambris JD, Wells AD, Song WC. The complement inhibitory protein DAF (CD55) suppresses T cell immunity in vivo. J Exp Med 2005;201:567-577.
  39. Fridkis-Hareli M, Storek M, Or E, Altman R, Katti S, Sun F, Peng T, Hunter J, Johnson K, Wang Y, et al. The human complement receptor type 2 (CR2)/CR1 fusion protein TT32, a novel targeted inhibitor of the classical and alternative pathway C3 convertases, prevents arthritis in active immunization and passive transfer mouse models. Mol Immunol 2019;105:150-164.
  40. Ozen A, Comrie WA, Ardy RC, Dominguez Conde C, Dalgic B, Beser OF, Morawski AR, Karakoc-Aydiner E, Tutar E, Baris S, et al. CD55 deficiency, early-onset protein-losing enteropathy, and thrombosis. N Engl J Med 2017;377:52-61.
  41. Egan ES, Jiang RH, Moechtar MA, Barteneva NS, Weekes MP, Nobre LV, Gygi SP, Paulo JA, Frantzreb C, Tani Y, et al. Malaria. A forward genetic screen identifies erythrocyte CD55 as essential for Plasmodium falciparum invasion. Science 2015;348:711-714.
  42. Thurman JM, Nester CM. All things complement. Clin J Am Soc Nephrol 2016;11:1856-1866.
  43. Li Q, Huang D, Nacion K, Bu H, Lin F. Augmenting DAF levels in vivo ameliorates experimental autoimmune encephalomyelitis. Mol Immunol 2009;46:2885-2891.
  44. Hamann J, Wishaupt JO, van Lier RA, Smeets TJ, Breedveld FC, Tak PP. Expression of the activation antigen CD97 and its ligand CD55 in rheumatoid synovial tissue. Arthritis Rheum 1999;42:650-658.
  45. Hill A, DeZern AE, Kinoshita T, Brodsky RA. Paroxysmal nocturnal haemoglobinuria. Nat Rev Dis Primers 2017;3:17028.
  46. van den Wijngaard RM, Asghar SS, Pijnenborg AC, Tigges AJ, Westerhof W, Das PK. Aberrant expression of complement regulatory proteins, membrane cofactor protein and decay accelerating factor, in the involved epidermis of patients with vitiligo. Br J Dermatol 2002;146:80-87.
  47. Qiao P, Dang EL, Fang H, Zhang JY, Li B, Shen SX, Luo YX, Lei J, Shao S, Qiao HJ, et al. Decreased expression levels of complement regulator CD55 contribute to the development of bullous pemphigoid. Oncotarget 2018;9:35517-35527.
  48. Bharti R, Dey G, Lin F, Lathia J, Reizes O. CD55 in cancer: complementing functions in a non-canonical manner. Cancer Lett 2022;551:215935.