Knee Osteoarthritis: A Review of Pathogenesis and State-Of-The-Art Non-Operative Therapeutic Considerations

Dragan Primorac, Vilim Molnar, Eduard Rod, Željko Jeleč, Fabijan Čukelj, Vid Matišić, Trpimir Vrdoljak, Damir Hudetz, Hana Hajsok, Igor Borić, Dragan Primorac, Vilim Molnar, Eduard Rod, Željko Jeleč, Fabijan Čukelj, Vid Matišić, Trpimir Vrdoljak, Damir Hudetz, Hana Hajsok, Igor Borić

Abstract

Being the most common musculoskeletal progressive condition, osteoarthritis is an interesting target for research. It is estimated that the prevalence of knee osteoarthritis (OA) among adults 60 years of age or older is approximately 10% in men and 13% in women, making knee OA one of the leading causes of disability in elderly population. Today, we know that osteoarthritis is not a disease characterized by loss of cartilage due to mechanical loading only, but a condition that affects all of the tissues in the joint, causing detectable changes in tissue architecture, its metabolism and function. All of these changes are mediated by a complex and not yet fully researched interplay of proinflammatory and anti-inflammatory cytokines, chemokines, growth factors and adipokines, all of which can be measured in the serum, synovium and histological samples, potentially serving as biomarkers of disease stage and progression. Another key aspect of disease progression is the epigenome that regulates all the genetic expression through DNA methylation, histone modifications, and mRNA interference. A lot of work has been put into developing non-surgical treatment options to slow down the natural course of osteoarthritis to postpone, or maybe even replace extensive surgeries such as total knee arthroplasty. At the moment, biological treatments such as platelet-rich plasma, bone marrow mesenchymal stem cells and autologous microfragmented adipose tissue containing stromal vascular fraction are ordinarily used. Furthermore, the latter two mentioned cell-based treatment options seem to be the only methods so far that increase the quality of cartilage in osteoarthritis patients. Yet, in the future, gene therapy could potentially become an option for orthopedic patients. In the following review, we summarized all of the latest and most important research in basic sciences, pathogenesis, and non-operative treatment.

Keywords: cytokines; epigenomics; genetic therapy; knee osteoarthritis; mesenchymal stem cells; phenotype; platelet-rich plasma.

Conflict of interest statement

The authors declare no conflict of interest.

Figures

Figure 1
Figure 1
Schematic representation of cartilage extracellular matrix and its changes in osteoarthritis. (A) A healthy network of proteoglycan aggregates entangled with type II collagen fibres is seen. matrix metalloproteinases (MMPs) and a disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS) cleavage sites are represented by red arrows; (B) Cartilage matrix changes in osteoarthritis defined by degradation of proteoglycans and cleavage of type II collagen fibres by ADAMTS and MMPs, respectively.
Figure 2
Figure 2
Microarchitectural and histologic changes of articular cartilage, subchondral bone and synovium in osteoarthritis. (A) Representation of normal joint structure and normal histologic state of the main tissue involved in osteoarthritis; (B) Early-stage osteoarthritis changes in articular cartilage, subchondral bone and synovium. Shallow cartilage erosions of the superficial tangential zone and middle transitional zone causing disruption of the extracellular matrix can be seen. Secondary chondrocyte hypertrophy and clustering is present as well. Changes to other joint tissues, mainly the reduction in subchondral bone mass, synovial thickening and inflammatory cells (lymphocytes) migration, together with changes in cellular activity and count, are depicted. (C) Late-stage osteoarthritis changes of involved tissues. Full-thickness cartilage erosions reaching the subchondral bone, chondrocyte apoptosis, subchondral bone sclerosis, osteophyte and subchondral cysts formation alongside vascular infiltration are shown. Further synovial thickening with immune cells infiltration and increased vascularization is also seen.

References

    1. Bortoluzzi A., Furini F., Scirè C.A. Osteoarthritis and its management—Epidemiology, nutritional aspects and environmental factors. Autoimmun. Rev. 2018;17:1097–1104. doi: 10.1016/j.autrev.2018.06.002.
    1. Mabey T., Honsawek S. Cytokines as biochemical markers for knee osteoarthritis. World J. Orthop. 2015;6:95–105. doi: 10.5312/wjo.v6.i1.95.
    1. Nelson A.E. Osteoarthritis year in review 2017: Clinical. Osteoarthr. Cartil. 2018;26:319–325. doi: 10.1016/j.joca.2017.11.014.
    1. Loeser R.F., Goldring S.R., Scanzello C.R., Goldring M.B. Osteoarthritis: A disease of the joint as an organ. Arthritis Rheum. 2012;64:1697–1707. doi: 10.1002/art.34453.
    1. 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;2:1–18. doi: 10.1038/nrdp.2016.72.
    1. Swingler T.E., Niu L., Smith P., Paddy P., Le L., Barter M.J., Young D.A., Clark I.M. The function of microRNAs in cartilage and osteoarthritis. Clin. Exp. Rheumatol. 2019;37:40–47.
    1. Ilas D.C., Churchman S.M., McGonagle D., Jones E. Targeting subchondral bone mesenchymal stem cell activities for intrinsic joint repair in osteoarthritis. Futur. Sci. OA. 2017;3:FSO228. doi: 10.4155/fsoa-2017-0055.
    1. Hunter D.J., Bierma-Zeinstra S. Osteoarthritis. Lancet. 2019;393:1745–1759. doi: 10.1016/S0140-6736(19)30417-9.
    1. Carlson A.K., Rawle R.A., Wallace C.W., Brooks E.G., Adams E., Greenwood M.C., Olmer M., Lotz M.K., Bothner B., June R.K. Characterization of synovial fluid metabolomic phenotypes of cartilage morphological changes associated with osteoarthritis. Osteoarthr. Cartil. 2019;27:1174–1184. doi: 10.1016/j.joca.2019.04.007.
    1. Nguyen U.-S.D.T., Zhang Y., Zhu Y., Niu J., Zhang B., Felson D.T. Increasing Prevalence of Knee Pain and Symptomatic Knee Osteoarthritis: Survey and Cohort Data. Ann. Intern. Med. 2011;155:725–732. doi: 10.7326/0003-4819-155-11-201112060-00004.
    1. Zhang Y., Jordan J.M. Epidemiology of Osteoarthritis. Clin. Geriatr. Med. 2010;26:355–369. doi: 10.1016/j.cger.2010.03.001.
    1. GBD 2015 Disease and Injury Incidence and Prevalence Collaborators Global, regional, and national incidence, prevalence, and years lived with disability for 310 diseases and injuries, 1990–2015: A systematic analysis for the Global Burden of Disease Study 2015. Lancet. 2016;388:1545–1602. doi: 10.1016/S0140-6736(16)31678-6.
    1. Prince M.J., Wu F., Guo Y., Gutierrez Robledo L.M., O’Donnell M., Sullivan R., Yusuf S. The burden of disease in older people and implications for health policy and practice. Lancet. 2015;385:549–562. doi: 10.1016/S0140-6736(14)61347-7.
    1. Shane Anderson A., Loeser R.F. Why is osteoarthritis an age-related disease? Best Pract. Res. Clin. Rheumatol. 2010;24:15–26. doi: 10.1016/j.berh.2009.08.006.
    1. Felson D.T. Osteoarthritis as a disease of mechanics. Osteoarthr. Cartil. 2013;21:10–15. doi: 10.1016/j.joca.2012.09.012.
    1. Vina E.R., Kwoh C.K. Epidemiology of osteoarthritis. Curr. Opin. Rheumatol. 2018;30:160–167. doi: 10.1097/BOR.0000000000000479.
    1. Harris E.C., Coggon D. HIP osteoarthritis and work. Best Pract. Res. Clin. Rheumatol. 2015;29:462–482. doi: 10.1016/j.berh.2015.04.015.
    1. Ezzat A.M., Li L.C. Occupational Physical Loading Tasks and Knee Osteoarthritis: A Review of the Evidence. Physiother. Can. 2014;66:91–107. doi: 10.3138/ptc.2012-45BC.
    1. Driban J.B., Hootman J.M., Sitler M.R., Harris K.P., Cattano N.M. Is Participation in Certain Sports Associated With Knee Osteoarthritis? A Systematic Review. J. Athl. Train. 2017;52:497–506. doi: 10.4085/1062-6050-50.2.08.
    1. Berenbaum F., Wallace I.J., Lieberman D.E., Felson D.T. Modern-day environmental factors in the pathogenesis of osteoarthritis. Nat. Rev. Rheumatol. 2018;14:674–681. doi: 10.1038/s41584-018-0073-x.
    1. Hame S.L., Alexander R.A. Knee osteoarthritis in women. Curr. Rev. Musculoskelet. Med. 2013;6:182–187. doi: 10.1007/s12178-013-9164-0.
    1. Long H., Zeng X., Liu Q., Wang H., Vos T., Hou Y., Lin C., Qiu Y., Wang K., Xing D., et al. Burden of osteoarthritis in China, 1990–2017: Findings from the Global Burden of Disease Study 2017. Lancet Rheumatol. 2020;2:e164–e172. doi: 10.1016/S2665-9913(19)30145-6.
    1. Wang H., Bai J., He B., Hu X., Liu D. Osteoarthritis and the risk of cardiovascular disease: A meta-analysis of observational studies. Sci. Rep. 2016;6:1–7. doi: 10.1038/srep39672.
    1. Wallace I.J., Worthington S., Felson D.T., Jurmain R.D., Wren K.T., Maijanen H., Woods R.J., Lieberman D.E. Knee osteoarthritis has doubled in prevalence since the mid-20th century. Proc. Natl. Acad. Sci. USA. 2017;114:9332–9336. doi: 10.1073/pnas.1703856114.
    1. Veronese N., Cereda E., Maggi S., Luchini C., Solmi M., Smith T., Denkinger M., Hurley M., Thompson T., Manzato E., et al. Osteoarthritis and mortality: A prospective cohort study and systematic review with meta-analysis. Semin. Arthritis Rheum. 2016;46:160–167. doi: 10.1016/j.semarthrit.2016.04.002.
    1. Bianco D., Todorov A., Čengić T., Pagenstert G., Schären S., Netzer C., Hügle T., Geurts J. Alterations of Subchondral Bone Progenitor Cells in Human Knee and Hip Osteoarthritis Lead to a Bone Sclerosis Phenotype. Int. J. Mol. Sci. 2018;19:475. doi: 10.3390/ijms19020475.
    1. Reynard L.N., Barter M.J. Osteoarthritis year in review 2019: Genetics, genomics and epigenetics. Osteoarthr. Cartil. 2020;28:275–284. doi: 10.1016/j.joca.2019.11.010.
    1. Rice S.J., Beier F., Young D.A., Loughlin J. Interplay between genetics and epigenetics in osteoarthritis. Nat. Rev. Rheumatol. 2020;16:268–281. doi: 10.1038/s41584-020-0407-3.
    1. Emery C.A., Whittaker J.L., Mahmoudian A., Lohmander L.S., Roos E.M., Bennell K.L., Toomey C.M., Reimer R.A., Thompson D., Ronsky J.L., et al. Establishing outcome measures in early knee osteoarthritis. Nat. Rev. Rheumatol. 2019;15:438–448. doi: 10.1038/s41584-019-0237-3.
    1. Xia B., Chen D., Zhang J., Hu S., Jin H., Tong P. Osteoarthritis Pathogenesis: A Review of Molecular Mechanisms. Calcif. Tissue Int. 2014;95:495–505. doi: 10.1007/s00223-014-9917-9.
    1. Goldring S.R., Goldring M.B. Changes in the osteochondral unit during osteoarthritis: Structure, function and cartilage bone crosstalk. Nat. Rev. Rheumatol. 2016;12:632–644. doi: 10.1038/nrrheum.2016.148.
    1. Carballo C.B., Nakagawa Y., Sekiya I., Rodeo S.A. Basic Science of Articular Cartilage. Clin. Sports Med. 2017;36:413–425. doi: 10.1016/j.csm.2017.02.001.
    1. Primorac D., Stover M.L., Clark S.H., Rowe D.W. Molecular basis of nanomelia, a heritable chondrodystrophy of chicken. Matrix Biol. 1994;14:297–305. doi: 10.1016/0945-053X(94)90195-3.
    1. Primorac D. Reduced Type II collagen mRNA in nanomelic cultured chondrocytes: An example of extracellular matrix/collagen feedback regulation? Croat. Med. J. 1995;36:85–92.
    1. Primorac D., Johnson C., Lawrence J., McKinstry M., Stover M., Schanfield M., Andelinović S., Tadić T., Rowe D. Premature termination codon in the aggrecan gene of nanomelia and its influence on mRNA transport and stability. Croat. Med. J. 1999;40:528–532.
    1. Houard X., Goldring M.B., Berenbaum F. Homeostatic Mechanisms in Articular Cartilage and Role of Inflammation in Osteoarthritis. Curr. Rheumatol. Rep. 2013;15:375. doi: 10.1007/s11926-013-0375-6.
    1. Goldring M.B., Marcu K.B. Cartilage homeostasis in health and rheumatic diseases. Arthritis Res. Ther. 2009;11:224. doi: 10.1186/ar2592.
    1. Musumeci G. The Effect of Mechanical Loading on Articular Cartilage. J. Funct. Morphol. Kinesiol. 2016;1:154–161. doi: 10.3390/jfmk1020154.
    1. Jacobs C.R., Huang H., Kwon R.Y. Introduction to Cell Mechanics and Mechanobiology. Garland Science; New York, NY, USA: 2012.
    1. Vanwanseele B., Eckstein F., Knecht H., Spaepen A., Stüssis E. Longitudinal Analysis of Cartilage Atrophy in the Knees of Patients with Spinal Cord Injury. Arthritis Rheum. 2003;48:3377–3381. doi: 10.1002/art.11367.
    1. Mansfield J.C., Mandalia V., Toms A., Winlove C.P., Brasselet S. Collagen reorganization in cartilage under strain probed by polarization sensitive second harmonic generation microscopy. J. R. Soc. Interface. 2019;16:20180611. doi: 10.1098/rsif.2018.0611.
    1. Mansfield J.C., Bell J.S., Winlove C.P. The micromechanics of the superficial zone of articular cartilage. Osteoarthr. Cartil. 2015;23:1806–1816. doi: 10.1016/j.joca.2015.05.030.
    1. Korhonen R.K., Julkunen P., Wilson W., Herzog W. Importance of Collagen Orientation and Depth-Dependent Fixed Charge Densities of Cartilage on Mechanical Behavior of Chondrocytes. J. Biomech. Eng. 2008;130:021003. doi: 10.1115/1.2898725.
    1. Wilson W., Driessen N.J.B., van Donkelaar C.C., Ito K. Prediction of collagen orientation in articular cartilage by a collagen remodeling algorithm. Osteoarthr. Cartil. 2006;14:1196–1202. doi: 10.1016/j.joca.2006.05.006.
    1. Wu J.P., Kirk T.B., Zheng M.H. Study of the collagen structure in the superficial zone and physiological state of articular cartilage using a 3D confocal imaging technique. J. Orthop. Surg. Res. 2008;3:29. doi: 10.1186/1749-799X-3-29.
    1. Mansfield J.C., Peter Winlove C. A multi-modal multiphoton investigation of microstructure in the deep zone and calcified cartilage. J. Anat. 2012;220:405–416. doi: 10.1111/j.1469-7580.2012.01479.x.
    1. Ruhlen R., Marberry K. The chondrocyte primary cilium. Osteoarthr. Cartil. 2014;22:1071–1076. doi: 10.1016/j.joca.2014.05.011.
    1. Goldring M.B., Goldring S.R. Articular cartilage and subchondral bone in the pathogenesis of osteoarthritis. Ann. N. Y Acad. Sci. 2010;1192:230–237. doi: 10.1111/j.1749-6632.2009.05240.x.
    1. Stanton H., Rogerson F.M., East C.J., Golub S.B., Lawlor K.E., Meeker C.T., Little C.B., Last K., Farmer P.J., Campbell I.K., et al. ADAMTS5 is the major aggrecanase in mouse cartilage in vivo and in vitro. Nature. 2005;434:648–652. doi: 10.1038/nature03417.
    1. Pratta M.A., Su J.L., Leesnitzer M.A., Struglics A., Larsson S., Lohmander L.S., Kumar S. Development and characterization of a highly specific and sensitive sandwich ELISA for detection of aggrecanase-generated aggrecan fragments. Osteoarthr. Cartil. 2006;14:702–713. doi: 10.1016/j.joca.2006.01.012.
    1. Loeser R.F. Molecular mechanisms of cartilage destruction: Mechanics, inflammatory mediators, and aging collide. Arthritis Rheum. 2006;54:1357–1360. doi: 10.1002/art.21813.
    1. Parrish A.R. Matrix Metalloproteinases and Tissue Remodeling in Health and Disease: Target Tissues and Therapy. 1st ed. Elsevier; San Diego, CA, USA: 2017. p. 310.
    1. Rolauffs B., Williams J.M., Aurich M., Grodzinsky A.J., Kuettner K.E., Cole A.A. Proliferative Remodeling of the Spatial Organization of Human Superficial Chondrocytes Distant From Focal Early Osteoarthritis. Arthritis Rheum. Off. J. Am. Coll. Rheum. 2010;62:489–498. doi: 10.1002/art.27217.
    1. Glyn-Jones S., Palmer A.J.R., Agricola R., Price A.J., Vincent T.L., Weinans H., Carr A.J. Osteoarthritis. Lancet. 2015;386:376–387. doi: 10.1016/S0140-6736(14)60802-3.
    1. Zhen G., Cao X. Targeting TGFβ signaling in subchondral bone and articular cartilage homeostasis. Trends Pharmacol. Sci. 2014;35:227–236. doi: 10.1016/j.tips.2014.03.005.
    1. Li G., Yin J., Gao J., Cheng T.S., Pavlos N.J., Zhang C., Zheng M.H. Subchondral bone in osteoarthritis: Insight into risk factors and microstructural changes. Arthritis Res. Ther. 2013;15:223. doi: 10.1186/ar4405.
    1. Funck-Brentano T., Cohen-Solal M. Subchondral bone and osteoarthritis. Curr. Opin. Rheumatol. 2015;27:420–426. doi: 10.1097/BOR.0000000000000181.
    1. Sanchez C., Pesesse L., Gabay O., Delcour J.P., Msika P., Baudouin C., Henrotin Y.E. Regulation of subchondral bone osteoblast metabolism by cyclic compression. Arthritis Rheum. 2012;64:1193–1203. doi: 10.1002/art.33445.
    1. Burr D.B., Gallant M.A. Bone remodelling in osteoarthritis. Nat. Rev. Rheumatol. 2012;8:665–673. doi: 10.1038/nrrheum.2012.130.
    1. Goldring S.R. Role of Bone in Osteoarthritis Pathogenesis. Med. Clin. N. Am. 2009;93:25–35. doi: 10.1016/j.mcna.2008.09.006.
    1. Driban J.B., Tassinari A., Lo G.H., Price L.L., Schneider E., Lynch J.A., Eaton C.B., McAlindon T.E. Bone marrow lesions are associated with altered trabecular morphometry. Osteoarthr. Cartil. 2012;20:1519–1526. doi: 10.1016/j.joca.2012.08.013.
    1. Bowes M.A., McLure S.W., Wolstenholme C.B., Vincent G.R., Williams S., Grainger A., Conaghan P.G. Osteoarthritic bone marrow lesions almost exclusively colocate with denuded cartilage: A 3D study using data from the Osteoarthritis Initiative. Ann. Rheum. Dis. 2016;75:1852–1857. doi: 10.1136/annrheumdis-2015-208407.
    1. Cotofana S., Wyman B.T.T., Benichou O., Dreher D., Nevitt M., Gardiner J., Wirth W., Hitzl W., Kwoh C.K.K., Eckstein F., et al. Relationship between knee pain and the presence, location, size and phenotype offemorotibial denuded areas of subchondral bone as visualized by MRI. Osteoarthr. Cartil. 2013;21:1214–1222. doi: 10.1016/j.joca.2013.04.001.
    1. Crema M.D., Roemer F.W., Zhu Y., Marra M.D., Niu J., Zhang Y., Lynch J.A., Javaid M.K., Lewis C.E., El-Khoury G.Y., et al. Subchondral cystlike lesions develop longitudinally in areas of bone marrow edema-like lesions in patients with or at risk for knee osteoarthritis: Detection with MR imaging—The MOST study. Radiology. 2010;256:855–862. doi: 10.1148/radiol.10091467.
    1. Yang Y., Li P., Zhu S., Bi R. Comparison of early-stage changes of osteoarthritis in cartilage and subchondral bone between two different rat models. PeerJ. 2020;8:e8934. doi: 10.7717/peerj.8934.
    1. Crema M.D.D., Cibere J., Sayre E.C.C., Roemer F.W.W., Wong H., Thorne A., Singer J., Esdaile J.M.M., Marra M.D.D., Kopec J.A.A., et al. The relationship between subchondral sclerosis detected with MRI and cartilage loss in a cohort of subjects with knee pain: The knee osteoarthritis progression (KOAP) study. Osteoarthr. Cartil. 2014;22:540–546. doi: 10.1016/j.joca.2014.01.006.
    1. Wenham C.Y.J., Conaghan P.G. The role of synovitis in osteoarthritis. Ther. Adv. Musculoskelet. Dis. 2010;2:349–359. doi: 10.1177/1759720X10378373.
    1. Schmidt T.A., Gastelum N.S., Nguyen Q.T., Schumacher B.L., Sah R.L. Boundary lubrication of articular cartilage: Role of synovial fluid constituents. Arthritis Rheum. 2007;56:882–891. doi: 10.1002/art.22446.
    1. Griffin T.M., Scanzello C.R. Innate inflammation and synovial macrophages in osteoarthritis pathophysiology. Clin. Exp. Rheumatol. 2019;37:57–63.
    1. 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;2019:1–18. doi: 10.1155/2019/6390182.
    1. Sellam J., Berenbaum F. The role of synovitis in pathophysiology and clinical symptoms of osteoarthritis. Nat. Rev. Rheumatol. 2010;6:625–635. doi: 10.1038/nrrheum.2010.159.
    1. Xie J., Huang Z., Yu X., Zhou L., Pei F. Clinical implications of macrophage dysfunction in the development of osteoarthritis of the knee. Cytokine Growth Factor Rev. 2019;46:36–44. doi: 10.1016/j.cytogfr.2019.03.004.
    1. HüGle T., Geurts J. What drives osteoarthritis?-synovial versus subchondral bone pathology. Rheumatology. 2016;56:1461–1471. doi: 10.1093/rheumatology/kew389.
    1. Ene R., Diana Sinescu R., Ene P., Mihaela Cîrstoiu M., Cătălin Cîrstoiu F. Synovial inflammation in patients with different stages of knee osteoarthritis. Rom. J. Morphol. Embryol. 2015;56:169–173.
    1. de Lange-Brokaar B.J.E., Ioan-Facsinay A., van Osch G.J.V.M., Zuurmond A.-M., Schoones J., Toes R.E.M., Huizinga T.W.J., Kloppenburg M. Synovial inflammation, immune cells and their cytokines in osteoarthritis: A review. Osteoarthr. Cartil. 2012;20:1484–1499. doi: 10.1016/j.joca.2012.08.027.
    1. Ayral X., Pickering E.H., Woodworth T.G., Mackillop N., Dougados M. Synovitis: A potential predictive factor of structural progression of medial tibiofemoral knee osteoarthritis - Results of a 1 year longitudinal arthroscopic study in 422 patients. Osteoarthr. Cartil. 2005;13:361–367. doi: 10.1016/j.joca.2005.01.005.
    1. Kapoor M., Martel-Pelletier J., Lajeunesse D., Pelletier J.P., Fahmi H. Role of proinflammatory cytokines in the pathophysiology of osteoarthritis. Nat. Rev. Rheumatol. 2011;7:33–42. doi: 10.1038/nrrheum.2010.196.
    1. Yang F., Zhou S., Wang C., Huang Y., Li H., Wang Y., Zhu Z., Tang J., Yan M. Epigenetic modifications of interleukin-6 in synovial fibroblasts from osteoarthritis patients. Sci. Rep. 2017;7:1–11. doi: 10.1038/srep43592.
    1. Baker K., Grainger A., Niu J., Clancy M., Guermazi A., Crema M., Hughes L., Buckwalter J., Wooley A., Nevitt M., et al. Relation of synovitis to knee pain using contrast-enhanced MRIs. Ann. Rheum. Dis. 2010;69:1779–1783. doi: 10.1136/ard.2009.121426.
    1. Roemer F.W., Kassim Javaid M., Guermazi A., Thomas M., Kiran A., Keen R., King L., Arden N.K. Anatomical distribution of synovitis in knee osteoarthritis and its association with joint effusion assessed on non-enhanced and contrast-enhanced MRI. Osteoarthr. Cartil. 2010;18:1269–1274. doi: 10.1016/j.joca.2010.07.008.
    1. O’Neill T.W., Parkes M.J., Maricar N., Marjanovic E.J., Hodgson R., Gait A.D., Cootes T.F., Hutchinson C.E., Felson D.T. Synovial tissue volume: A treatment target in knee osteoarthritis (OA) Ann. Rheum. Dis. 2016;75:84–90. doi: 10.1136/annrheumdis-2014-206927.
    1. Hunter D.J., McDougall J.J., Keefe F.J. The Symptoms of Osteoarthritis and the Genesis of Pain. Rheum. Dis. Clin. N. Am. 2008;34:623–643. doi: 10.1016/j.rdc.2008.05.004.
    1. Fusco M., Skaper S.D., Coaccioli S., Varrassi G., Paladini A. Degenerative Joint Diseases and Neuroinflammation. Pain. Pract. 2017;17:522–532. doi: 10.1111/papr.12551.
    1. D’Agostino M.A., Conaghan P., Le Bars M., Baron G., Grassi W., Martin-Mola E., Wakefield R., Brasseur J.L., So A., Backhaus M., et al. EULAR report on the use of ultrasonography in painful knee osteoarthritis. Part 1: Prevalence of inflammation in osteoarthritis. Ann. Rheum. Dis. 2005;64:1703–1709. doi: 10.1136/ard.2005.037994.
    1. Fernandez-Madrid F., Karvonen R.L., Teitge R.A., Miller P.R., An T., Negendank W.G. Synovial thickening detected by MR imaging in osteoarthritis of the knee confirmed by biopsy as synovitis. Magn. Reson. Imaging. 1995;13:177–183. doi: 10.1016/0730-725X(94)00119-N.
    1. Loeuille D., Chary-Valckenaere I., Champigneulle J., Rat A.C., Toussaint F., Pinzano-Watrin A., Goebel J.C., Mainard D., Blum A., Pourel J., et al. Macroscopic and microscopic features of synovial membrane inflammation in the osteoarthritic knee: Correlating magnetic resonance imaging findings with disease severity. Arthritis Rheum. 2005;52:3492–3501. doi: 10.1002/art.21373.
    1. Eymard F., Chevalier X. Inflammation of the infrapatellar fat pad. Jt. Bone Spine. 2016;83:389–393. doi: 10.1016/j.jbspin.2016.02.016.
    1. Paduszyński W., Jeśkiewicz M., Uchański P., Gackowski S., Radkowski M., Demkow U. Advances in Experimental Medicine and Biology. Volume 1039. Springer; New York, NY, USA: 2017. Hoffa’s Fat Pad Abnormality in the Development of Knee Osteoarthritis; pp. 95–102.
    1. Roemer F.W., Jarraya M., Felson D.T., Hayashi D., Crema M.D., Loeuille D., Guermazi A. Magnetic resonance imaging of Hoffa’s fat pad and relevance for osteoarthritis research: A narrative review. Osteoarthr. Cartil. 2016;24:383–397. doi: 10.1016/j.joca.2015.09.018.
    1. Barboza E., Hudson J., Chang W.P., Kovats S., Towner R.A., Silasi-Mansat R., Lupu F., Kent C., Griffin T.M. Profibrotic Infrapatellar Fat Pad Remodeling Without M1 Macrophage Polarization Precedes Knee Osteoarthritis in Mice With Diet-Induced Obesity. Arthritis Rheumatol. 2017;69:1221–1232. doi: 10.1002/art.40056.
    1. Wu C.L., Harasymowicz N.S., Klimak M.A., Collins K.H., Guilak F. The role of macrophages in osteoarthritis and cartilage repair. Osteoarthr. Cartil. 2020;28:544–554. doi: 10.1016/j.joca.2019.12.007.
    1. Ushiyama T., Chano T., Inoue K., Matsusue Y. Cytokine production in the infrapatellar fat pad: Another source of cytokines in knee synovial fluids. Ann. Rheum. Dis. 2003;62:108–112. doi: 10.1136/ard.62.2.108.
    1. Simopoulou T., Malizos K.N., Iliopoulos D., Stefanou N., Papatheodorou L., Ioannou M., Tsezou A. Differential expression of leptin and leptin’s receptor isoform (Ob-Rb) mRNA between advanced and minimally affected osteoarthritic cartilage; effect on cartilage metabolism. Osteoarthr. Cartil. 2007;15:872–883. doi: 10.1016/j.joca.2007.01.018.
    1. Englund M., Roemer F.W., Hayashi D., Crema M.D., Guermazi A. Meniscus pathology, osteoarthritis and the treatment controversy. Nat. Rev. Rheumatol. 2012;8:412–419. doi: 10.1038/nrrheum.2012.69.
    1. Hunter D.J., Zhang Y.Q., Niu J.B., Tu X., Amin S., Clancy M., Guermazi A., Grigorian M., Gale D., Felson D.T. The association of meniscal pathologic changes with cartilage loss in symptomatic knee osteoarthritis. Arthritis Rheum. 2006;54:795–801. doi: 10.1002/art.21724.
    1. Ding C., Martel-Pelletier J., Pelletier J.P., Abram F., Raynauld J.P., Cicuttini F., Jones G. Meniscal tear as an osteoarthritis risk factor in a largely non-osteoarthritic cohort: A cross-sectional study. J. Rheumatol. 2007;34:776–784.
    1. Crema M.D., Guermazi A., Li L., Nogueira-Barbosa M.H., Marra M.D., Roemer F.W., Eckstein F., Hellio Le Graverand M.P., Wyman B.T., Hunter D.J. The association of prevalent medial meniscal pathology with cartilage loss in the medial tibiofemoral compartment over a 2-year period. Osteoarthr. Cartil. 2010;18:336–343. doi: 10.1016/j.joca.2009.11.003.
    1. Englund M. The role of biomechanics in the initiation and progression of OA of the knee. Best Pract. Res. Clin. Rheumatol. 2010;24:39–46. doi: 10.1016/j.berh.2009.08.008.
    1. Roemer F.W., Guermazi A., Hunter D.J., Niu J., Zhang Y., Englund M., Javaid M.K., Lynch J.A., Mohr A., Torner J., et al. The association of meniscal damage with joint effusion in persons without radiographic osteoarthritis: The Framingham and MOST osteoarthritis studies. Osteoarthr. Cartil. 2009;17:748–753. doi: 10.1016/j.joca.2008.09.013.
    1. Crema M.D., Roemer F.W., Felson D.T., Englund M., Wang K., Jarraya M., Nevitt M.C., Marra M.D., Torner J.C., Lewis C.E., et al. Factors associated with meniscal extrusion in knees with or at risk for osteoarthritis: The multicenter osteoarthritis study. Radiology. 2012;264:494–503. doi: 10.1148/radiol.12110986.
    1. Stehling C., Souza R.B., Le Graverand M.P.H., Wyman B.T., Li X., Majumdar S., Link T.M. Loading of the knee during 3.0 T MRI is associated with significantly increased medial meniscus extrusion in mild and moderate osteoarthritis. Eur. J. Radiol. 2012;81:1839–1845. doi: 10.1016/j.ejrad.2011.05.027.
    1. van der Voet J.A., Runhaar J., van der Plas P., Vroegindeweij D., Oei E.H., Bierma-Zeinstra S.M.A. Baseline meniscal extrusion associated with incident knee osteoarthritis after 30 months in overweight and obese women. Osteoarthr. Cartil. 2017;25:1299–1303. doi: 10.1016/j.joca.2017.03.014.
    1. Liikavainio T., Lyytinen T., Tyrväinen E., Sipilä S., Arokoski J.P. Physical Function and Properties of Quadriceps Femoris Muscle in Men With Knee Osteoarthritis. Arch. Phys. Med. Rehabil. 2008;89:2185–2194. doi: 10.1016/j.apmr.2008.04.012.
    1. Alnahdi A.H., Zeni J.A., Snyder-Mackler L. Muscle impairments in patients with knee osteoarthritis. Sports Health. 2012;4:284–292. doi: 10.1177/1941738112445726.
    1. Kim J.-R., Yoo J., Kim H. Therapeutics in Osteoarthritis Based on an Understanding of Its Molecular Pathogenesis. Int. J. Mol. Sci. 2018;19:674. doi: 10.3390/ijms19030674.
    1. Roos E.M., Herzog W., Block J.A., Bennell K.L. Muscle weakness, afferent sensory dysfunction and exercise in knee osteoarthritis. Nat. Rev. Rheumatol. 2011;7:57–63. doi: 10.1038/nrrheum.2010.195.
    1. Raynauld J.-P., Pelletier J.-P., Roubille C., Dorais M., Abram F., Li W., Wang Y., Fairley J., Cicuttini F.M., Martel-Pelletier J. Magnetic Resonance Imaging-Assessed Vastus Medialis Muscle Fat Content and Risk for Knee Osteoarthritis Progression: Relevance From a Clinical Trial. Arthritis Care Res. 2015;67:1406–1415. doi: 10.1002/acr.22590.
    1. Teichtahl A.J., Wluka A.E., Wang Y., Wijethilake P.N., Strauss B.J., Proietto J., Dixon J.B., Jones G., Forbes A., Cicuttini F.M. Vastus medialis fat infiltration—A modifiable determinant of knee cartilage loss. Osteoarthr. Cartil. 2015;23:2150–2157. doi: 10.1016/j.joca.2015.06.016.
    1. Krishnasamy P., Hall M., Robbins S.R. The role of skeletal muscle in the pathophysiology and management of knee osteoarthritis. Rheumatology. 2018;57:iv22–iv33. doi: 10.1093/rheumatology/kex515.
    1. Rothrauff B.B., Jorge A., de Sa D., Kay J., Fu F.H., Musahl V. Anatomic ACL reconstruction reduces risk of post-traumatic osteoarthritis: A systematic review with minimum 10-year follow-up. Knee Surg. Sport Traumatol. Arthrosc. 2019;28:1072–1084. doi: 10.1007/s00167-019-05665-2.
    1. Wang Y., Wluka A.E., Berry P.A., Siew T., Teichtahl A.J., Urquhart D.M., Lloyd D.G., Jones G., Cicuttini F.M. Increase in vastus medialis cross-sectional area is associated with reduced pain, cartilage loss, and joint replacement risk in knee osteoarthritis. Arthritis Rheum. 2012;64:3917–3925. doi: 10.1002/art.34681.
    1. Mobasheri A., Henrotin Y. Biomarkers of (osteo)arthritis. Biomarkers. 2015;20:513–518. doi: 10.3109/1354750X.2016.1140930.
    1. Boehme K.A., Rolauffs B. Onset and Progression of Human Osteoarthritis—Can Growth Factors, Inflammatory Cytokines, or Differential miRNA Expression Concomitantly Induce Proliferation, ECM Degradation, and Inflammation in Articular Cartilage? Int. J. Mol. Sci. 2018;19:2282. doi: 10.3390/ijms19082282.
    1. Goldring M.B., Goldring S.R. Osteoarthritis. J. Cell. Physiol. 2007;213:626–634. doi: 10.1002/jcp.21258.
    1. Wojdasiewicz P., Poniatowski Ł.A., Szukiewicz D. The Role of Inflammatory and Anti-Inflammatory Cytokines in the Pathogenesis of Osteoarthritis. Mediat. Inflamm. 2014;2014:1–19. doi: 10.1155/2014/561459.
    1. Mathiessen A., Conaghan P.G. Synovitis in osteoarthritis: Current understanding with therapeutic implications. Arthritis Res. Ther. 2017;19:1–9. doi: 10.1186/s13075-017-1229-9.
    1. Nguyen L., Sharma A., Chakraborty C., Saibaba B., Ahn M.-E., Lee S.-S. Review of Prospects of Biological Fluid Biomarkers in Osteoarthritis. Int. J. Mol. Sci. 2017;18:601. doi: 10.3390/ijms18030601.
    1. Mabey T., Honsawek S., Tanavalee A., Yuktanandana P., Wilairatana V., Poovorawan Y. Plasma and synovial fluid inflammatory cytokine profiles in primary knee osteoarthritis. Biomarkers. 2016;21:639–644. doi: 10.3109/1354750X.2016.1171907.
    1. Zhu Z., Otahal P., Wang B., Jin X., Laslett L.L.L., Wluka A.E.E., Antony B., Han W., Wang X., Winzenberg T., et al. Cross-sectional and longitudinal associations between serum inflammatory cytokines and knee bone marrow lesions in patients with knee osteoarthritis. Osteoarthr. Cartil. 2017;25:499–505. doi: 10.1016/j.joca.2016.10.024.
    1. Yang P., Tan J., Yuan Z., Meng G., Bi L., Liu J. Expression profile of cytokines and chemokines in osteoarthritis patients: Proinflammatory roles for CXCL8 and CXCL11 to chondrocytes. Int. Immunopharmacol. 2016;40:16–23. doi: 10.1016/j.intimp.2016.08.005.
    1. Halász K., Kassner A., Mörgelin M., Heinegård D. COMP acts as a catalyst in collagen fibrillogenesis. J. Biol. Chem. 2007;282:31166–31173. doi: 10.1074/jbc.M705735200.
    1. Ruan G., Xu J., Wang K., Wu J., Zhu Q., Ren J., Bian F., Chang B., Bai X., Han W., et al. Associations between knee structural measures, circulating inflammatory factors and MMP13 in patients with knee osteoarthritis. Osteoarthr. Cartil. 2018;26:1063–1069. doi: 10.1016/j.joca.2018.05.003.
    1. Goldring M.B., Berenbaum F. The Regulation of Chondrocyte Function by Proinflammatory Mediators. Clin. Orthop. Relat. Res. 2004;427:S37–S46. doi: 10.1097/01.blo.0000144484.69656.e4.
    1. Yang Q., Zhou Y., Cai P., Fu W., Wang J., Wei Q., Li X. Up-regulated HIF-2α contributes to the Osteoarthritis development through mediating the primary cilia loss. Int. Immunopharmacol. 2019;75:105762. doi: 10.1016/j.intimp.2019.105762.
    1. Alaaeddine N., Olee T., Hashimoto S., Creighton-Achermann L., Lotz M. Production of the chemokine RANTES by articular chondrocytes and role in cartilage degradation. Arthritis Rheum. 2001;44:1633–1643. doi: 10.1002/1529-0131(200107)44:7<1633::AID-ART286>;2-Z.
    1. Akeson G., Malemud C. A Role for Soluble IL-6 Receptor in Osteoarthritis. J. Funct. Morphol. Kinesiol. 2017;2:27. doi: 10.3390/jfmk2030027.
    1. Latourte A., Cherifi C., Maillet J., Ea H.K., Bouaziz W., Funck-Brentano T., Cohen-Solal M., Hay E., Richette P. Systemic inhibition of IL-6/Stat3 signalling protects against experimental osteoarthritis. Ann. Rheum. Dis. 2017;76:748–755. doi: 10.1136/annrheumdis-2016-209757.
    1. Qu X., Wang W.J., Tang S., Liu Y., Wang J. Correlation between interleukin-6 expression in articular cartilage bone and osteoarthritis. Genet. Mol. Res. 2015;14:14189–14195. doi: 10.4238/2015.November.13.2.
    1. Pearson M.J., Herndler-Brandstetter D., Tariq M.A., Nicholson T.A., Philp A.M., Smith H.L., Davis E.T., Jones S.W., Lord J.M. IL-6 secretion in osteoarthritis patients is mediated by chondrocyte-synovial fibroblast cross-talk and is enhanced by obesity. Sci. Rep. 2017;7:3451. doi: 10.1038/s41598-017-03759-w.
    1. Heinegård D., Saxne T. The role of the cartilage matrix in osteoarthritis. Nat. Rev. Rheumatol. 2011;7:50–56. doi: 10.1038/nrrheum.2010.198.
    1. Giordano R., Petersen K.K., Andersen H.H., Simonsen O., Arendt-Nielsen L. Serum Inflammatory Markers in Patients with Knee Osteoarthritis: A Proteomic Approach. Clin. J. Pain. 2020;36:229–237. doi: 10.1097/AJP.0000000000000804.
    1. Larsson S., Lohmander L.S., Struglics A. Synovial fluid level of aggrecan ARGS fragments is a more sensitive marker of joint disease than glycosaminoglycan or aggrecan levels: A cross-sectional study. Arthritis Res. Ther. 2009;11:R92. doi: 10.1186/ar2735.
    1. Zhou J.L., Fang H.S., Peng H., Deng S., Chen S., Li J.P., Qiu B., Weng J.Q., Liu F. The relationship between HIF-2α and VEGF with radiographic severity in the primary osteoarthritic knee. Yonsei Med. J. 2016;57:735–740. doi: 10.3349/ymj.2016.57.3.735.
    1. Venkatesan J.K., Rey-Rico A., Schmitt G., Wezel A., Madry H., Cucchiarini M. rAAV-mediated overexpression of TGF-β stably restructures human osteoarthritic articular cartilage in situ. J. Transl. Med. 2013;11:211. doi: 10.1186/1479-5876-11-211.
    1. Carballo C.B., Coelho T.R.P., de Holanda Afonso R.C., de O. Faria J.C., Alves T., Monte S.M., Ventura Matioszek G.M., Moura-Neto V., de Brito J.M. Osteoarthritic Synovial Fluid and TGF-β1 Induce Interleukin-18 in Articular Chondrocytes. Cartilage. 2018:194760351879614. doi: 10.1177/1947603518796149.
    1. Ruiz M., Toupet K., Maumus M., Rozier P., Jorgensen C., Noël D. TGFBI secreted by mesenchymal stromal cells ameliorates osteoarthritis and is detected in extracellular vesicles. Biomaterials. 2020;226:119544. doi: 10.1016/j.biomaterials.2019.119544.
    1. Zhen G., Wen C., Jia X., Li Y., Crane J.L., Mears S.C., Askin F.B., Frassica F.J., Chang W., Yao J., et al. Inhibition of TGF-β signaling in mesenchymal stem cells of subchondral bone attenuates osteoarthritis. Nat. Med. 2013;19:704–712. doi: 10.1038/nm.3143.
    1. Waly N.E., Refaiy A., Aborehab N.M. IL-10 and TGF-β: Roles in chondroprotective effects of Glucosamine in experimental Osteoarthritis? Pathophysiology. 2017;24:45–49. doi: 10.1016/j.pathophys.2017.02.005.
    1. Heard B.J., Rosvold J.M., Fritzler M.J., El-Gabalawy H., Wiley J.P., Krawetz R.J. A computational method to differentiate normal individuals, osteoarthritis and rheumatoid arthritis patients using serum biomarkers. J. R. Soc. Interface. 2014;11:20140428. doi: 10.1098/rsif.2014.0428.
    1. Kisand K., Tamm A.E., Lintrop M., Tamm A.O. New insights into the natural course of knee osteoarthritis: Early regulation of cytokines and growth factors, with emphasis on sex-dependent angiogenesis and tissue remodeling. A pilot study. Osteoarthr. Cartil. 2018;26:1045–1054. doi: 10.1016/j.joca.2018.05.009.
    1. Leung Y.Y., Huebner J.L., Haaland B., Wong S.B.S., Kraus V.B. Synovial fluid pro-inflammatory profile differs according to the characteristics of knee pain. Osteoarthr. Cartil. 2017;25:1420–1427. doi: 10.1016/j.joca.2017.04.001.
    1. Nees T.A., Rosshirt N., Zhang J.A., Reiner T., Sorbi R., Tripel E., Walker T., Schiltenwolf M., Hagmann S., Moradi B. Synovial Cytokines Significantly Correlate with Osteoarthritis-Related Knee Pain and Disability: Inflammatory Mediators of Potential Clinical Relevance. J. Clin. Med. 2019;8:1343. doi: 10.3390/jcm8091343.
    1. Grieshaber-Bouyer R., Kämmerer T., Rosshirt N., Nees T.A., Koniezke P., Tripel E., Schiltenwolf M., Kirsch J., Hagmann S., Moradi B. Divergent Mononuclear Cell Participation and Cytokine Release Profiles Define Hip andKnee Osteoarthritis. J. Clin. Med. 2019;8:1631. doi: 10.3390/jcm8101631.
    1. Ren G., Lutz I., Railton P., Wiley J.P., McAllister J., Powell J., Krawetz R.J. Serum and synovial fluid cytokine profiling in hip osteoarthritis: Distinct from knee osteoarthritis and correlated with pain. BMC Musculoskelet. Disord. 2018;19:1–11. doi: 10.1186/s12891-018-1955-4.
    1. WHO | Obesity. [(accessed on 4 May 2020)]; Available online:
    1. Blüher M. Obesity: Global epidemiology and pathogenesis. Nat. Rev. Endocrinol. 2019;15:288–298. doi: 10.1038/s41574-019-0176-8.
    1. Murphy L., Schwartz T.A., Helmick C.G., Renner J.B., Tudor G., Koch G., Dragomir A., Kalsbeek W.D., Luta G., Jordan J.M. Lifetime risk of symptomatic knee osteoarthritis. Arthritis Care Res. 2008;59:1207–1213. doi: 10.1002/art.24021.
    1. Powell A. Obesity: A preventable risk factor for large joint osteoarthritis which may act through biomechanical factors. Br. J. Sports Med. 2005;39:4–5. doi: 10.1136/bjsm.2004.011841.
    1. Aspden R.M. Obesity punches above its weight in osteoarthritis. Nat. Rev. Rheumatol. 2011;7:65–68. doi: 10.1038/nrrheum.2010.123.
    1. Sharma L., Song J., Felson D.T., Cahue S., Shamiyeh E., Dunlop D.D. The role of knee alignment in disease progression and functional decline in knee osteoarthritis. J. Am. Med. Assoc. 2001;286:188–195. doi: 10.1001/jama.286.2.188.
    1. Urban H., Little C.B. The role of fat and inflammation in the pathogenesis and management of osteoarthritis. Rheumatology. 2018;57:iv10–iv21. doi: 10.1093/rheumatology/kex399.
    1. Kluzek S., Arden N.K., Newton J. Adipokines as potential prognostic biomarkers in patients with acute knee injury. Biomarkers. 2015;20:519–525. doi: 10.3109/1354750X.2014.948914.
    1. Collins A.T., Kulvaranon M.L., Cutcliffe H.C., Utturkar G.M., Smith W.A.R.R., Spritzer C.E., Guilak F., DeFrate L.E. Obesity alters the in vivo mechanical response and biochemical properties of cartilage as measured by MRI. Arthritis Res. Ther. 2018;20:232. doi: 10.1186/s13075-018-1727-4.
    1. Boyce L., Prasad A., Barrett M., Dawson-Bowling S., Millington S., Hanna S.A., Achan P. The outcomes of total knee arthroplasty in morbidly obese patients: A systematic review of the literature. Arch. Orthop. Trauma Surg. 2019;139:553–560. doi: 10.1007/s00402-019-03127-5.
    1. Choi Y.R., Collins K.H., Lee J.W., Kang H.J., Guilak F. Genome Engineering for Osteoarthritis: From Designer Cells to Disease-Modifying Drugs. Tissue Eng. Regen. Med. 2019;16:335–343. doi: 10.1007/s13770-018-0172-4.
    1. Shen J., Wang C., Li D., Xu T., Myers J., Ashton J.M., Wang T., Zuscik M.J., McAlinden A., O’Keefe R.J. DNA methyltransferase 3b regulates articular cartilage homeostasis by altering metabolism. JCI Insight. 2017;2:e93612. doi: 10.1172/jci.insight.93612.
    1. Cao K., Wei L., Zhang Z., Guo L., Zhang C., Li Y., Sun C., Sun X., Wang S., Li P., et al. Decreased histone deacetylase 4 is associated with human osteoarthritis cartilage degeneration by releasing histone deacetylase 4 inhibition of runt-related transcription factor-2 and increasing osteoarthritis-related genes: A novel mechanism of human ost. Arthritis Res. Ther. 2014;16:491. doi: 10.1186/s13075-014-0491-3.
    1. Richard D., Liu Z., Cao J., Kiapour A.M., Willen J., Yarlagadda S., Jagoda E., Kolachalama V.B., Sieker J.T., Chang G.H., et al. Evolutionary Selection and Constraint on Human Knee Chondrocyte Regulation Impacts Osteoarthritis Risk. Cell. 2020;181:362–381. doi: 10.1016/j.cell.2020.02.057.
    1. Miyamoto Y., Mabuchi A., Shi D., Kubo T., Takatori Y., Saito S., Fujioka M., Sudo A., Uchida A., Yamamoto S., et al. A functional polymorphism in the 5′ UTR of GDF5 is associated with susceptibility to osteoarthritis. Nat. Genet. 2007;39:529–533. doi: 10.1038/2005.
    1. Zengini E., Hatzikotoulas K., Tachmazidou I., Steinberg J., Hartwig F.P., Southam L., Hackinger S., Boer C.G., Styrkarsdottir U., Gilly A., et al. Genome-wide analyses using UK Biobank data provide insights into the genetic architecture of osteoarthritis. Nat. Genet. 2018;50:549–558. doi: 10.1038/s41588-018-0079-y.
    1. Liu Y., Chang J.-C., Hon C.-C., Fukui N., Tanaka N., Zhang Z., Lee M.T.M., Minoda A. Chromatin accessibility landscape of articular knee cartilage reveals aberrant enhancer regulation in osteoarthritis. Sci. Rep. 2018;8:15499. doi: 10.1038/s41598-018-33779-z.
    1. Yu X.-M., Meng H.-Y., Yuan X.-L., Wang Y., Guo Q.-Y., Peng J., Wang A.-Y., Lu S.-B. MicroRNAs’ Involvement in Osteoarthritis and the Prospects for Treatments. Evid. Based Complement. Altern. Med. 2015;2015:1–13. doi: 10.1155/2015/236179.
    1. Zhang M., Lygrissea K., Wanga J. Role of MicroRNA in Osteoarthritis. J. Arthritis. 2017;06:239. doi: 10.4172/2167-7921.1000239.
    1. Zhang X., Wang C., Zhao J., Xu J., Geng Y., Dai L., Huang Y., Fu S.C., Dai K., Zhang X. MiR-146a facilitates osteoarthritis by regulating cartilage homeostasis via targeting Camk2d and Ppp3r2. Cell Death Dis. 2017;8:1–11. doi: 10.1038/cddis.2017.146.
    1. Coutinho De Almeida R., Ramos Y.F.M., Mahfouz A., Den Hollander W., Lakenberg N., Houtman E., Van Hoolwerff M., Rodríguez Ruiz A., Slagboom P.E., Mei H., et al. TranslaTional science RNA sequencing data integration reveals an miRNA interactome of osteoarthritis cartilage. Ann. Rheum. Dis. 2019;78:270–277. doi: 10.1136/annrheumdis-2018-213882.
    1. Miyaki S., Asahara H. Macro view of microRNA function in osteoarthritis. Nat. Rev. Rheumatol. 2012;8:543–552. doi: 10.1038/nrrheum.2012.128.
    1. Grigelioniene G., Suzuki H.I., Taylan F., Mirzamohammadi F., Borochowitz Z.U., Ayturk U.M., Tzur S., Horemuzova E., Lindstrand A., Weis M.A., et al. Gain-of-function mutation of microRNA-140 in human skeletal dysplasia. Nat. Med. 2019;25:583–590. doi: 10.1038/s41591-019-0353-2.
    1. Hu S., Zhao X., Mao G., Zhang Z.Z., Wen X., Zhang C., Liao W., Zhang Z.Z. MicroRNA-455-3p promotes TGF-β signaling and inhibits osteoarthritis development by directly targeting PAK2. Exp. Mol. Med. 2019;51:1–13. doi: 10.1038/s12276-019-0322-3.
    1. Hu G., Zhao X., Wang C., Geng Y., Zhao J., Xu J., Zuo B., Zhao C., Wang C., Zhang X. MicroRNA-145 attenuates TNF-α-driven cartilage matrix degradation in osteoarthritis via direct suppression of MKK4. Cell Death Dis. 2017;8:e3140. doi: 10.1038/cddis.2017.522.
    1. Ragni E., Perucca Orfei C., De Luca P., Viganò M., Colombini A., Lugano G., Bollati V., de Girolamo L. miR-22-5p and miR-29a-5p Are Reliable Reference Genes for Analyzing Extracellular Vesicle-Associated miRNAs in Adipose-Derived Mesenchymal Stem Cells and Are Stable under Inflammatory Priming Mimicking Osteoarthritis Condition. Stem Cell Rev. Rep. 2019;15:743–754. doi: 10.1007/s12015-019-09899-y.
    1. Vicente R., Noël D., Pers Y.M., Apparailly F., Jorgensen C. Deregulation and therapeutic potential of microRNAs in arthritic diseases. Nat. Rev. Rheumatol. 2016;12:211–220. doi: 10.1038/nrrheum.2015.162.
    1. Huang J., Zhao L., Fan Y., Liao L., Ma P.X., Xiao G., Chen D. The microRNAs miR-204 and miR-211 maintain joint homeostasis and protect against osteoarthritis progression. Nat. Commun. 2019;10:1–13. doi: 10.1038/s41467-019-10753-5.
    1. Kang D., Shin J., Cho Y., Kim H.-S., Gu Y.-R., Kim H., You K.T., Chang M.J., Chang C.B., Kang S.-B., et al. Stress-activated miR-204 governs senescent phenotypes of chondrocytes to promote osteoarthritis development. Sci. Transl. Med. 2019;11:eaar6659. doi: 10.1126/scitranslmed.aar6659.
    1. Bianchi M., Renzini A., Adamo S., Moresi V. Coordinated Actions of MicroRNAs with other Epigenetic Factors Regulate Skeletal Muscle Development and Adaptation. Int. J. Mol. Sci. 2017;18:840. doi: 10.3390/ijms18040840.
    1. Grover A.K., Samson S.E. Benefits of antioxidant supplements for knee osteoarthritis: Rationale and reality. Nutr. J. 2015;15:1. doi: 10.1186/s12937-015-0115-z.
    1. Blanco F.J., Rego I., Ruiz-Romero C. The role of mitochondria in osteoarthritis. Nat. Rev. Rheumatol. 2011;7:161–169. doi: 10.1038/nrrheum.2010.213.
    1. Rego-Pérez I., Durán-Sotuela A., Ramos-Louro P., Blanco F.J. Mitochondrial Genetics and Epigenetics in Osteoarthritis. Front. Genet. 2020;10:1335. doi: 10.3389/fgene.2019.01335.
    1. Andia I., Maffulli N. Platelet-rich plasma for managing pain and inflammation in osteoarthritis. Nat. Rev. Rheumatol. 2013;9:721–730. doi: 10.1038/nrrheum.2013.141.
    1. Dohan Ehrenfest D.M., Andia I., Zumstein M.A., Zhang C.-Q., Pinto N.R., Bielecki T. Classification of platelet concentrates (Platelet-Rich Plasma-PRP, Platelet-Rich Fibrin-PRF) for topical and infiltrative use in orthopedic and sports medicine: Current consensus, clinical implications and perspectives. Muscle Ligaments Tendons J. 2019;4:3–9. doi: 10.32098/mltj.01.2014.02.
    1. Wu Q., Luo X., Xiong Y., Liu G., Wang J., Chen X., Mi B. Platelet-rich plasma versus hyaluronic acid in knee osteoarthritis: A meta-analysis with the consistent ratio of injection. J. Orthop. Surg. 2020;28:1–9. doi: 10.1177/2309499019887660.
    1. Gato-Calvo L., Magalhaes J., Ruiz-Romero C., Blanco F.J., Burguera E.F. Platelet-rich plasma in osteoarthritis treatment: Review of current evidence. Ther. Adv. Chronic Dis. 2019;10:1–18. doi: 10.1177/2040622319825567.
    1. Sundaram K., Vargas-Hernández J.S., Sanchez T.R., Moreu N.M., Mont M.A., Higuera C.A., Piuzzi N.S. Are Subchondral Intraosseous Injections Effective and Safe for the Treatment of Knee Osteoarthritis? A Systematic Review. J. Knee Surg. 2019;32:1046–1057. doi: 10.1055/s-0039-1677792.
    1. Sánchez M., Delgado D., Pompei O., Pérez J.C., Sánchez P., Garate A., Bilbao A.M., Fiz N., Padilla S. Treating Severe Knee Osteoarthritis with Combination of Intra-Osseous and Intra-Articular Infiltrations of Platelet-Rich Plasma: An Observational Study. Cartilage. 2019;10:245–253. doi: 10.1177/1947603518756462.
    1. Ponchel F., Burska A.N., Hensor E.M.A., Raja R., Campbell M., Emery P., Conaghan P.G. Changes in peripheral blood immune cell composition in osteoarthritis. Osteoarthr. Cartil. 2015;23:1870–1878. doi: 10.1016/j.joca.2015.06.018.
    1. Daghestani H.N., Kraus V.B. Inflammatory biomarkers in osteoarthritis. Osteoarthr. Cartil. 2015;23:1890–1896. doi: 10.1016/j.joca.2015.02.009.
    1. Caplan A.I. Medicinal signalling cells: They work, so use them. Nature. 2019;566:39. doi: 10.1038/d41586-019-00490-6.
    1. Caplan A.I., Hariri R. Body Management: Mesenchymal Stem Cells Control the Internal Regenerator. Stem Cells Transl. Med. 2015;4:695–701. doi: 10.5966/sctm.2014-0291.
    1. Caplan A.I. What’s in a Name? Tissue Eng. Part A. 2010;16:2415–2417. doi: 10.1089/ten.tea.2010.0216.
    1. McGonagle D., Baboolal T.G., Jones E. Native joint-resident mesenchymal stem cells for cartilage repair in osteoarthritis. Nat. Rev. Rheumatol. 2017;13:719–730. doi: 10.1038/nrrheum.2017.182.
    1. De Windt T.S., Saris D.B.F., Slaper-Cortenbach I.C.M., Van Rijen M.H.P., Gawlitta D., Creemers L.B., De Weger R.A., Dhert W.J.A., Vonk L.A. Direct cell-cell contact with chondrocytes is a key mechanism in multipotent mesenchymal stromal cell-mediated chondrogenesis. Tissue Eng. Part A. 2015;21:2536–2547. doi: 10.1089/ten.tea.2014.0673.
    1. Mancuso P., Raman S., Glynn A., Barry F., Murphy J.M. Mesenchymal Stem Cell Therapy for Osteoarthritis: The Critical Role of the Cell Secretome. Front. Bioeng. Biotechnol. 2019;7:9. doi: 10.3389/fbioe.2019.00009.
    1. Pittenger M.F., Discher D.E., Péault B.M., Phinney D.G., Hare J.M., Caplan A.I. Mesenchymal stem cell perspective: Cell biology to clinical progress. Npj Regen. Med. 2019;4:22. doi: 10.1038/s41536-019-0083-6.
    1. Ren G., Zhang L., Zhao X., Xu G., Zhang Y., Roberts A.I., Zhao R.C., Shi Y. Mesenchymal Stem Cell-Mediated Immunosuppression Occurs via Concerted Action of Chemokines and Nitric Oxide. Cell Stem Cell. 2008;2:141–150. doi: 10.1016/j.stem.2007.11.014.
    1. Wang Y., Yu D., Liu Z., Zhou F., Dai J., Wu B., Zhou J., Heng B.C., Zou X.H., Ouyang H., et al. Exosomes from embryonic mesenchymal stem cells alleviate osteoarthritis through balancing synthesis and degradation of cartilage extracellular matrix. Stem Cell Res. Ther. 2017;8:189. doi: 10.1186/s13287-017-0632-0.
    1. Kim S.H., Ha C.-W., Park Y.-B., Nam E., Lee J.-E., Lee H.-J. Intra-articular injection of mesenchymal stem cells for clinical outcomes and cartilage repair in osteoarthritis of the knee: A meta-analysis of randomized controlled trials. Arch. Orthop. Trauma Surg. 2019;139:971–980. doi: 10.1007/s00402-019-03140-8.
    1. Caplan A.I., Correa D. The MSC: An Injury Drugstore. Cell Stem Cell. 2011;9:11–15. doi: 10.1016/j.stem.2011.06.008.
    1. Hass R., Kasper C., Böhm S., Jacobs R. Different populations and sources of human mesenchymal stem cells (MSC): A comparison of adult and neonatal tissue-derived MSC. Cell Commun. Signal. 2011;9:12. doi: 10.1186/1478-811X-9-12.
    1. Hassan H.T., El-Sheemy M. Adult bone-marrow stem cells and their potential in medicine. J. R. Soc. Med. 2004;97:465–471. doi: 10.1177/0141076809701003.
    1. Chahal J., Gómez-Aristizábal A., Shestopaloff K., Bhatt S., Chaboureau A., Fazio A., Chisholm J., Weston A., Chiovitti J., Keating A., et al. Bone Marrow Mesenchymal Stromal Cell Treatment in Patients with Osteoarthritis Results in Overall Improvement in Pain and Symptoms and Reduces Synovial Inflammation. Stem Cells Transl. Med. 2019;8:746–757. doi: 10.1002/sctm.18-0183.
    1. Gupta P.K., Chullikana A., Rengasamy M., Shetty N., Pandey V., Agarwal V., Wagh S.Y., Vellotare P.K., Damodaran D., Viswanathan P., et al. Efficacy and safety of adult human bone marrow-derived, cultured, pooled, allogeneic mesenchymal stromal cells (Stempeucel®): Preclinical and clinical trial in osteoarthritis of the knee joint. Arthritis Res. Ther. 2016;18:301. doi: 10.1186/s13075-016-1195-7.
    1. Vega A., Martín-Ferrero M.A., Del Canto F., Alberca M., García V., Munar A., Orozco L., Soler R., Fuertes J.J., Huguet M., et al. Treatment of knee osteoarthritis with allogeneic bone marrow mesenchymal stem cells: A randomized controlled trial. Transplantation. 2015;99:1681–1690. doi: 10.1097/TP.0000000000000678.
    1. Awad M.E., Hussein K.A., Helwa I., Abdelsamid M.F., Aguilar-Perez A., Mohsen I., Hunter M., Hamrick M.W., Isales C.M., Elsalanty M., et al. Meta-Analysis and Evidence Base for the Efficacy of Autologous Bone Marrow Mesenchymal Stem Cells in Knee Cartilage Repair: Methodological Guidelines and Quality Assessment. Stem Cells Int. 2019;2019:1–15. doi: 10.1155/2019/3826054.
    1. Maumus M., Pers Y.-M., Ruiz M., Jorgensen C., Noël D. Cellules souches mésenchymateuses et médecine régénératrice. Méd. Sci. 2018;34:1092–1099. doi: 10.1051/medsci/2018294.
    1. Hudetz D., Jeleč Ž., Rod E., Borić I., Plečko M., Primorac D. The Future of Cartilage Repair. In: Bodiroga-Vukobrat N., Rukavina D., Pavelić K., Sander G.G., editors. Personalized Medicine in Healthcare Systems: Legal, Medical and Economic Implications. Springer International Publishing; Cham, Switzerland: 2019. pp. 375–411.
    1. Hudetz D., Borić I., Rod E., Jeleč Ž., Radić A., Vrdoljak T., Skelin A., Lauc G., Trbojević-Akmačić I., Plečko M., et al. The Effect of Intra-articular Injection of Autologous Microfragmented Fat Tissue on Proteoglycan Synthesis in Patients with Knee Osteoarthritis. Genes. 2017;8:270. doi: 10.3390/genes8100270.
    1. Hudetz D., Borić I., Rod E., Jeleč Ž., Kunovac B., Polašek O., Vrdoljak T., Plečko M., Skelin A., Polančec D., et al. Early results of intra-articular micro-fragmented lipoaspirate treatment in patients with late stages knee osteoarthritis: A prospective study. Croat. Med. J. 2019;60:227–236. doi: 10.3325/cmj.2019.60.227.
    1. Polancec D., Zenic L., Hudetz D., Boric I., Jelec Z., Rod E., Vrdoljak T., Skelin A., Plecko M., Turkalj M., et al. Immunophenotyping of a Stromal Vascular Fraction from Microfragmented Lipoaspirate Used in Osteoarthritis Cartilage Treatment and Its Lipoaspirate Counterpart. Genes. 2019;10:474. doi: 10.3390/genes10060474.
    1. Borić I., Hudetz D., Rod E., Jeleč Ž., Vrdoljak T., Skelin A., Polašek O., Plečko M., Trbojević-Akmačić I., Lauc G., et al. A 24-Month Follow-Up Study of the Effect of Intra-Articular Injection of Autologous Microfragmented Fat Tissue on Proteoglycan Synthesis in Patients with Knee Osteoarthritis. Genes. 2019;10:1051. doi: 10.3390/genes10121051.
    1. Russo A., Screpis D., Di Donato S.L., Bonetti S., Piovan G., Zorzi C. Autologous micro-fragmented adipose tissue for the treatment of diffuse degenerative knee osteoarthritis: An update at 3 year follow-up. J. Exp. Orthop. 2018;5:52. doi: 10.1186/s40634-018-0169-x.
    1. Mautner K., Bowers R., Easley K., Fausel Z., Robinson R. Functional Outcomes Following Microfragmented Adipose Tissue Versus Bone Marrow Aspirate Concentrate Injections for Symptomatic Knee Osteoarthritis. Stem Cells Transl. Med. 2019;8:1149–1156. doi: 10.1002/sctm.18-0285.
    1. Russo A., Condello V., Madonna V., Guerriero M., Zorzi C. Autologous and micro-fragmented adipose tissue for the treatment of diffuse degenerative knee osteoarthritis. J. Exp. Orthop. 2017;4:33. doi: 10.1186/s40634-017-0108-2.
    1. Peretti G.M., Ulivi M., De Girolamo L., Meroni V., Lombardo M.D., Mangiavini L. Evaluation of the use of autologous micro-fragmented adipose tissue in the treatment of knee osteoarthritis: Preliminary results of a randomized controlled trial. J. Biol. Regul. Homeost. Agents. 2018;32:193–199.
    1. Freitag J., Bates D., Wickham J., Shah K., Huguenin L., Tenen A., Paterson K., Boyd R. Adipose-derived mesenchymal stem cell therapy in the treatment of knee osteoarthritis: A randomized controlled trial. Regen. Med. 2019;14:213–230. doi: 10.2217/rme-2018-0161.
    1. Yun S., Ku S.-K., Kwon Y.-S. Adipose-derived mesenchymal stem cells and platelet-rich plasma synergistically ameliorate the surgical-induced osteoarthritis in Beagle dogs. J. Orthop. Surg. Res. 2016;11:9. doi: 10.1186/s13018-016-0342-9.
    1. Pak J., Chang J.-J., Lee J.H., Lee S.H. Safety reporting on implantation of autologous adipose tissue-derived stem cells with platelet-rich plasma into human articular joints. BMC Musculoskelet. Disord. 2013;14:337. doi: 10.1186/1471-2474-14-337.
    1. Jayaram P., Ikpeama U., Rothenberg J.B., Malanga G.A. Bone Marrow-Derived and Adipose-Derived Mesenchymal Stem Cell Therapy in Primary Knee Osteoarthritis: A Narrative Review. PM&R. 2019;11:177–191. doi: 10.1016/j.pmrj.2018.06.019.
    1. Shariatzadeh M., Song J., Wilson S.L. The efficacy of different sources of mesenchymal stem cells for the treatment of knee osteoarthritis. Cell Tissue Res. 2019;378:399–410. doi: 10.1007/s00441-019-03069-9.
    1. Wu X., Wang Y., Xiao Y., Crawford R., Mao X., Prasadam I. Extracellular vesicles: Potential role in osteoarthritis regenerative medicine. J. Orthop. Transl. 2020;21:73–80. doi: 10.1016/j.jot.2019.10.012.
    1. Lener T., Gimona M., Aigner L., Börger V., Buzas E., Camussi G., Chaput N., Chatterjee D., Court F.A., del Portillo H.A., et al. Applying extracellular vesicles based therapeutics in clinical trials—An ISEV position paper. J. Extracell. Vesicles. 2015;4:30087. doi: 10.3402/jev.v4.30087.
    1. Morrison T.J., Jackson M.V., Cunningham E.K., Kissenpfennig A., McAuley D.F., O’Kane C.M., Krasnodembskaya A.D. Mesenchymal Stromal Cells Modulate Macrophages in Clinically Relevant Lung Injury Models by Extracellular Vesicle Mitochondrial Transfer. Am. J. Respir. Crit. Care Med. 2017;196:1275–1286. doi: 10.1164/rccm.201701-0170OC.
    1. Tan S.S.H., Tjio C.K.E., Wong J.R.Y., Wong K.L., Chew J.R.J., Hui J.H.P., Toh W.S. Mesenchymal Stem Cell Exosomes for Cartilage Regeneration: A Systematic Review of Preclinical In Vivo Studies. Tissue Eng. Part B Rev. 2020;2019:0326. doi: 10.1089/ten.teb.2019.0326.
    1. Sun Q., Zhang Y., Yang G., Chen X., Zhang Y., Cao G., Wang J., Sun Y., Zhang P., Fan M., et al. Transforming Growth Factor-Beta-Regulated miR-24 Promotes Skeletal Muscle Differentiation. Nucleic Acids Res. 2008;36:2690–2699. doi: 10.1093/nar/gkn032.
    1. Fleury A., Martinez M.C., Le Lay S. Extracellular Vesicles as Therapeutic Tools in Cardiovascular Diseases. Front. Immunol. 2014;5:370. doi: 10.3389/fimmu.2014.00370.
    1. Goldie B.J., Dun M.D., Lin M., Smith N.D., Verrills N.M., Dayas C.V., Cairns M.J. Activity-associated miRNA are packaged in Map1b-enriched exosomes released from depolarized neurons. Nucleic Acids Res. 2014;42:9195–9208. doi: 10.1093/nar/gku594.
    1. Dell’Isola A., Allan R., Smith S.L., Marreiros S.S.P., Steultjens M. Identification of clinical phenotypes in knee osteoarthritis: A systematic review of the literature. BMC Musculoskelet. Disord. 2016;17:1–12. doi: 10.1186/s12891-016-1286-2.
    1. Dell’Isola A., Steultjens M., Dell’Isola A., Steultjens M., Isola A.D., Steultjens M. Classification of patients with knee osteoarthritis in clinical phenotypes: Data from the osteoarthritis initiative. PLoS ONE. 2018;13:e191045. doi: 10.1371/journal.pone.0191045.
    1. Van Spil W.E., Kubassova O., Boesen M., Bay-Jensen A.-C., Mobasheri A. Osteoarthritis phenotypes and novel therapeutic targets. Biochem. Pharmacol. 2019;165:41–48. doi: 10.1016/j.bcp.2019.02.037.
    1. Chen T.-M., Chen Y.-H., Sun H., Tsai S.-J. Fibroblast growth factors: Potential novel targets for regenerative therapy of osteoarthritis. Chin. J. Physiol. 2019;62:2. doi: 10.4103/CJP.CJP_11_19.
    1. Meloni G., Farran A., Mohanraj B., Guehring H., Cocca R., Rabut E., Mauck R., Dodge G. Recombinant human FGF18 preserves depth-dependent mechanical inhomogeneity in articular cartilage. Eur. Cells Mater. 2019;38:23–38. doi: 10.22203/eCM.v038a03.
    1. Eckstein F., Kraines J.L., Aydemir A., Wirth W., Maschek S., Hochberg M.C. Intra-articular sprifermin reduces cartilage loss in addition to increasing cartilage gain independent of location in the femorotibial joint: Post-hoc analysis of a randomised, placebo-controlled phase II clinical trial. Ann. Rheum. Dis. 2020;79:525–528. doi: 10.1136/annrheumdis-2019-216453.
    1. Hochberg M.C., Guermazi A., Guehring H., Aydemir A., Wax S., Fleuranceau-Morel P., Reinstrup Bihlet A., Byrjalsen I., Ragnar Andersen J., Eckstein F. Effect of Intra-Articular Sprifermin vs Placebo on Femorotibial Joint Cartilage Thickness in Patients With Osteoarthritis. JAMA. 2019;322:1360. doi: 10.1001/jama.2019.14735.
    1. Gigout A., Guehring H., Froemel D., Meurer A., Ladel C., Reker D., Bay-Jensen A.C., Karsdal M.A., Lindemann S. Sprifermin (rhFGF18) enables proliferation of chondrocytes producing a hyaline cartilage matrix. Osteoarthr. Cartil. 2017;25:1858–1867. doi: 10.1016/j.joca.2017.08.004.
    1. Gregori D., Giacovelli G., Minto C., Barbetta B., Gualtieri F., Azzolina D., Vaghi P., Rovati L.C. Association of Pharmacological Treatments With Long-term Pain Control in Patients With Knee Osteoarthritis. JAMA. 2018;320:2564. doi: 10.1001/jama.2018.19319.
    1. Stöve J., Schneider-Wald B., Scharf H.P., Schwarz M.L. Bone morphogenetic protein 7 (bmp-7) stimulates Proteoglycan synthesis in human osteoarthritic chondrocytes in vitro. Biomed. Pharmacother. 2006;60:639–643. doi: 10.1016/j.biopha.2006.09.001.
    1. Bone morphogenetic protein 7 inhibits cartilage degradation in a rabbit model of osteoarthritis. Nat. Clin. Pract. Rheumatol. 2009;5:4. doi: 10.1038/ncprheum0955.
    1. Hayashi M., Muneta T., Ju Y.-J., Mochizuki T., Sekiya I. Weekly intra-articular injections of bone morphogenetic protein-7 inhibits osteoarthritis progression. Arthritis Res. Ther. 2008;10:R118. doi: 10.1186/ar2521.
    1. Hunter D.J., Pike M.C., Jonas B.L., Kissin E., Krop J., McAlindon T. Phase 1 safety and tolerability study of BMP-7 in symptomatic knee osteoarthritis. BMC Musculoskelet. Disord. 2010;11:232. doi: 10.1186/1471-2474-11-232.
    1. Mimpen J.Y., Snelling S.J.B. Chondroprotective Factors in Osteoarthritis: A Joint Affair. Curr. Rheumatol. Rep. 2019;21:1–14. doi: 10.1007/s11926-019-0840-y.
    1. Oo W.M., Yu S.P.C., Daniel M.S., Hunter D.J. Disease-modifying drugs in osteoarthritis: Current understanding and future therapeutics. Expert Opin. Emerg. Drugs. 2018;23:331–347. doi: 10.1080/14728214.2018.1547706.
    1. Shepard H.M., Phillips G.L., Thanos C.D., Feldmann M. Developments in therapy with monoclonal antibodies and related proteins. Clin. Med. J. R. Coll. Physicians Lond. 2017;17:220–232. doi: 10.7861/clinmedicine.17-3-220.
    1. Das V., Kc R., Li X., O-Sullivan I.S., van Wijnen A.J., Kroin J.S., Pytowski B., Applegate D.T., Votta-Velis G., Ripper R.L., et al. Blockade of vascular endothelial growth factor receptor-1 (Flt-1), reveals a novel analgesic for osteoarthritis-induced joint pain. Gene Rep. 2018;11:94–100. doi: 10.1016/j.genrep.2018.03.008.
    1. Kan S.-L., Li Y., Ning G.-Z., Yuan Z.-F., Chen L.-X., Bi M.-C., Sun J.-C., Feng S.-Q. Tanezumab for Patients with Osteoarthritis of the Knee: A Meta-Analysis. PLoS ONE. 2016;11:e0157105. doi: 10.1371/journal.pone.0157105.
    1. Schnitzer T.J., Easton R., Pang S., Levinson D.J., Pixton G., Viktrup L., Davignon I., Brown M.T., West C.R., Verburg K.M. Effect of Tanezumab on Joint Pain, Physical Function, and Patient Global Assessment of Osteoarthritis among Patients with Osteoarthritis of the Hip or Knee: A Randomized Clinical Trial. JAMA J. Am. Med. Assoc. 2019;322:37–48. doi: 10.1001/jama.2019.8044.
    1. Tiseo P.J., Kivitz A.J., Ervin J.E., Ren H., Mellis S.J. Fasinumab (REGN475), an antibody against nerve growth factor for the treatment of pain: Results from a double-blind, placebo-controlled exploratory study in osteoarthritis of the knee. Pain. 2014;155:1245–1252. doi: 10.1016/j.pain.2014.03.018.
    1. Gow J.M., Tsuji W.H., Williams G.J., Mytych D., Sciberras D., Searle S.L., Mant T., Gibbs J.P. Safety, tolerability, pharmacokinetics, and efficacy of AMG 403, a human anti-nerve growth factor monoclonal antibody, in two phase I studies with healthy volunteers and knee osteoarthritis subjects. Arthritis Res. Ther. 2015;17:282. doi: 10.1186/s13075-015-0797-9.
    1. Miller R.E., Tran P.B., Ishihara S., Larkin J., Malfait A.M. Therapeutic effects of an anti-ADAMTS-5 antibody on joint damage and mechanical allodynia in a murine model of osteoarthritis. Osteoarthr. Cartil. 2016;24:299–306. doi: 10.1016/j.joca.2015.09.005.
    1. Nixon A.J., Grol M.W., Lang H.M., Ruan M.Z.C., Stone A., Begum L., Chen Y., Dawson B., Gannon F., Plutizki S., et al. Disease-Modifying Osteoarthritis Treatment With Interleukin-1 Receptor Antagonist Gene Therapy in Small and Large Animal Models. Arthritis Rheumatol. 2018;70:1757–1768. doi: 10.1002/art.40668.
    1. Pickar-Oliver A., Gersbach C.A. The next generation of CRISPR–Cas technologies and applications. Nat. Rev. Mol. Cell Biol. 2019;20:490–507. doi: 10.1038/s41580-019-0131-5.
    1. Zhao L., Huang J., Fan Y., Li J., You T., He S., Xiao G., Chen D. Exploration of CRISPR/Cas9-based gene editing as therapy for osteoarthritis. Ann. Rheum. Dis. 2019;78:676–682. doi: 10.1136/annrheumdis-2018-214724.

Source: PubMed

3
Subscribe