Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 2428 2022-11-21 11:23:04 |
2 update references and layout Meta information modification 2428 2022-11-22 03:23:40 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Peshkova, M.;  Kosheleva, N.;  Shpichka, A.;  Radenska-Lopovok, S.;  Telyshev, D.;  Lychagin, A.;  Li, F.;  Timashev, P.;  Liang, X. Inflammation and Regeneration in Osteoarthritis. Encyclopedia. Available online: https://encyclopedia.pub/entry/35551 (accessed on 23 April 2024).
Peshkova M,  Kosheleva N,  Shpichka A,  Radenska-Lopovok S,  Telyshev D,  Lychagin A, et al. Inflammation and Regeneration in Osteoarthritis. Encyclopedia. Available at: https://encyclopedia.pub/entry/35551. Accessed April 23, 2024.
Peshkova, Maria, Nastasia Kosheleva, Anastasia Shpichka, Stefka Radenska-Lopovok, Dmitry Telyshev, Alexey Lychagin, Fangzhou Li, Peter Timashev, Xing-Jie Liang. "Inflammation and Regeneration in Osteoarthritis" Encyclopedia, https://encyclopedia.pub/entry/35551 (accessed April 23, 2024).
Peshkova, M.,  Kosheleva, N.,  Shpichka, A.,  Radenska-Lopovok, S.,  Telyshev, D.,  Lychagin, A.,  Li, F.,  Timashev, P., & Liang, X. (2022, November 21). Inflammation and Regeneration in Osteoarthritis. In Encyclopedia. https://encyclopedia.pub/entry/35551
Peshkova, Maria, et al. "Inflammation and Regeneration in Osteoarthritis." Encyclopedia. Web. 21 November, 2022.
Inflammation and Regeneration in Osteoarthritis
Edit

Osteoarthritis (OA) affects over 250 million people worldwide. It is a multifactorial disease characterized by cartilage loss and low-grade synovial inflammation. Focusing on these two targets together could be the key to developing currently missing disease-modifying OA drugs (DMOADs).

osteoarthritis inflammation synovitis cell-free tissue engineering

1. Introduction

Osteoarthritis (OA) has long been [1][2][3] and is sometimes still [4][5] referred to as a non-inflammatory condition. Moreover, when studying “classic” inflammatory arthritides, such as rheumatoid arthritis (RA) or spondyloarthritis, researchers would often use tissue and biologic fluid samples from OA patients as negative (non-inflammatory) controls [6][7]. Although currently researchers agree that many aspects contribute to OA progression, including gender [8], it has long been regarded mainly as the consequence of cartilage wear and tear. Cartilage damage was reported to lead to joint biomechanics impairment resulting in further cartilage loss and joint deformity, while the inflammatory component was commonly underestimated.
Cartilage tissue itself, being avascular, cannot develop a classic immune response. However, when considering the joint as a whole, including synovium, ligaments, and subchondral bone, inflammation (synovitis) seems to play an important role in OA progression. There is evidence that synovitis is associated with increased OA severity [9][10], and therefore it is a promising target for currently missing disease-modifying OA drug (DMOADs) development. Most available OA management options, including lifestyle changes, pharmacotherapy, and surgery [11][12][13], also target joint inflammation to varying degrees.
Lifestyle modification is the basis of most chronic disease management and can be beneficial to compliant patients. A balanced diet and physical activity can postpone the OA onset or slow down its progression both by reducing the joint loading due to weight loss and decreasing the levels of adipokines, which are known to contribute to the inflammatory component of OA development [14][15]. Currently recommended pharmacological treatments, including non-steroidal anti-inflammatory drugs (NSAIDs) and corticosteroids, target inflammation and relieve pain [11], while the operative approaches aim to both reduce pain and improve joint functions [13]. However, one of the major drawbacks of all these strategies is that none of them address the problem of cartilage loss. Due to its low self-healing capacity, hyaline cartilage can only be repaired with involvement of cartilage-repair techniques, and various strategies, both scaffold-based and scaffold-free and cell-based and cell-free, are being proposed [16][17]. Unfortunately, most conventional cartilage tissue repair techniques focus on cartilage regeneration, while their effect on joint inflammation is rarely being discussed. Apparently, in order to fully address the problem of OA, a combination of cartilage repair techniques and strategies targeting inflammation should be considered.

2. Inflammation in an OA Joint

Homeostasis in a healthy joint is maintained by the synovial intima cells, namely type A macrophage-like synoviocytes, responsible for debris phagocytosis, and type B fibroblast-like synoviocytes, which produce synovial fluid components, including hyaluronan [18]. In an OA joint the products of the cartilage extracellular matrix (ECM) degradation (thoroughly reviewed elsewhere [7]) are bound to the pattern recognition receptors (PRRs) and recognized by the innate immune system as damage/danger-associated molecular patterns (DAMPs). Other groups of DAMPs in an OA joint include plasma proteins (e.g., α1 and α2 microglobulins, fibrinogen, vitamin D-binding protein), crystals of basic calcium phosphate, calcium pyrophosphate dihydrate, and uric acid [19], and so-called alarmins [6], including HMGB1 and the S100 family of proteins. There is evidence of some DAMPs’ ability to activate the complement system [19]. Some DAMPs, for example, crystals, rather bind to cytoplasmic PRRs (e.g., NLRP3), initiating inflammasomes activation [20][21]. However, it seems that most DAMPs activate Toll-like receptors (TLRs), a large membrane-bound family of PRRs. TLRs are reported to be expressed both in the cartilage [22], being upregulated in the lesion areas [23], and in the synovium [19]. When stimulated by DAMPs, they trigger catabolic pathways in chondrocytes [23] and proinflammatory factor production by macrophages and mast cells [19]. The proinflammatory mediators can promote matrix metalloproteinases (MMP) production, directly enhancing the catabolic processes in the cartilage. They can also stimulate angiogenesis [24], increasing the influx of plasma proteins [7], which also act as DAMPs. Thus, more DAMPs are attracted to the area. Moreover, proinflammatory cytokines can boost their own production: exposure to proinflammatory cytokines promotes proinflammatory (M1) macrophage polarization, which in turn stimulates proinflammatory cytokine synthesis [25]. Thus, multiple vicious circles of further cartilage damage are established.
Synovial macrophages are the key cells orchestrating the processes of inflammation and healing within the joint because of their capacity to exhibit different phenotypes ranging from proinflammatory (M1) to anti-inflammatory (M2). In an OA joint, macrophages are caught into a vicious circle, being constantly attracted to the site by the perpetuated cartilage degradation and proinflammatory cytokine production, infiltrating the synovium and impairing its functions, notably, synovial fluid component synthesis [26], although to a lesser extent than in RA [27]. Alterations in the OA synovial fluid contents influence its properties, for example, the lack of hyaluronan leads to its decreased viscosity and elasticity [28], resulting in less effective lubrication of the joint surfaces and promoting cartilage damage. Aiming to find a way to disrupt the vicious circle involving the synovial macrophages, some researchers study the consequences of their depletion when modeling OA. Local macrophage depletion using intra-articular injections of clodronate-loaded liposomes has demonstrated rather positive outcomes in murine models, such as reduced MMPs’ expression in the synovial tissue and decreased osteophyte formation [29][30][31]. On the contrary, systemic depletion of macrophages in CSF-1R-GFP+ macrophage Fas-induced apoptosis (MaFIA)-transgenic mice placed on a high-fat diet induced systemic inflammation. Moreover, it led to massive infiltration of CD3+ T cells and neutrophils in the synovium [32], although neutrophil infiltration is characteristic of RA [33] and is hardly ever observed in OA [27].

3. Existing Strategies for Targeting Inflammation and Cartilage Regeneration in OA

3.1. Inflammation

OA can affect people of any age. Accidental trauma, for example, joint ligament damage, can lead to reactive inflammation and post-traumatic OA development. Even if the articular cartilage itself has not been damaged, the inflammatory process in the joint can lead to the activation of catabolic pathways in the cartilage tissue [34].
However, OA is generally discussed in the context of older adults and elderly patients, and one cannot talk about these groups of patients without mentioning comorbidities. Unfortunately, oral NSAIDs and acetaminophen (paracetamol), traditionally used for pain management in OA, are associated with adverse side effects. Those include gastrointestinal [35] and cardiovascular [36] damage for NSAIDs, while paracetamol use may be associated with liver [37] and renal [38] damage. On the other hand, topical NSAIDs are considered a safer but still rather effective option and are strongly recommended for OA patients [12]; however, their use is not enough to reverse the OA progression. Efficacy of intra-articular injections of steroids or hyaluronic acid (HA) remains debatable [11][12]. Mixed results were obtained when targeting proinflammatory cytokines as well. For example, anakinra, a recombinant human IL-1 receptor antagonist protein, was reported to perform better than placebo in patients with severe knee injury [39], and at the same time in OA patients its effect was comparable with placebo [40]. AMG 108, a monoclonal antibody binding the IL-1 receptor, also showed moderate effectiveness in OA patients with minimal clinical benefit [41]. Similarly, anti-tumor necrosis factor alpha (TNF-α) monoclonal antibodies, such as adalimumab and infliximab, despite sporadic encouraging evidence [42][43], were reported to have limited effectiveness in hand OA patients [44][45].
Speaking of arthroscopic procedures, joint lavage seems to hold promise in reducing inflammation following the removal of debris from the joint cavity; however, evidence suggests that it does not provide any significant improvement either [46].

3.2. Cartilage Regeneration

Despite undeniable progress in the field, cartilage regeneration remains a challenge. There are multiple approaches to cartilage repair depending on the size of the defect, and unfortunately each of them has some drawbacks. For example, bone marrow stimulating techniques, such as subchondral drilling and microfracturing, lead to the formation of fibrocartilage, whose mechanical properties are inferior to those of hyaline cartilage [47]. The use of bone marrow stimulation technique with hydrogel implantation into the defect provides the appropriate environment for hyaline cartilage formation [48]. Localized co-delivery of agents inducing hyaline cartilage formation such as bone morphogenic protein 2 (BMP2) and vascular endothelial growth factor (VEGF) receptor antagonist [49] in a hydrogel has also been proposed.
Another group of approaches is based on autologous chondrocyte implantation (ACI), sometimes applied together with collagen-based scaffolds (matrix-induced ACI, or MACI), and is used for larger cartilage defects. These approaches are reported to result in repair with hyaline-like cartilage; however, those are expensive multi-stage procedures requiring long-term rehabilitation [50].
In contrast, autologous stem cell transplantation may be performed in one stage, has a shorter rehabilitation period, and is less expensive than ACI. However, this approach requires longer-term studies to be recommended as a first-line treatment [50].
Osteochondral autografts or allografts are used for the largest cartilage lesions [51]. The osteochondral autograft transfer system (OATS, also known as mosaiplasty) has demonstrated good clinical outcomes, but even though the grafts are harvested from low-bearing regions in the joint, donor-site morbidity cannot be fully avoided. On the other hand, allografts, taken from deceased donors, seem to solve the problem of donor-site morbidity, but despite consensual cartilage immune privilege, some histocompatibility concerns cannot be ignored [52].

4. Inflammation Management in Tissue Engineering

In tissue engineering inflammation is mostly regarded as an adverse effect and a challenge to overcome. All the components of the tissue engineering triad (i.e., biomaterials, cells, and biochemical factors) are therefore being discussed in the context of biocompatibility. Furthermore, various modifications promoting better engraftment as well as minimizing the undesired immune reactions are being proposed for the existing approaches.

4.1. Biomaterials

Both natural and synthetic biomaterials used in tissue engineering have some strong advantages and some critical issues. For example, when assessing biomechanical properties or reproducibility, synthetic biomaterials, such as polycaprolactone (PCL), poly(glycolic acid) (PGA), polylactide (PLA), or poly(lactic-co-glycolic acid) (PLGA), are superior to natural biomaterials. On the other hand, when speaking about biocompatibility, natural biomaterials take precedence. It was reported that most synthetic polymers induce considerable inflammation in vivo [53][54], while natural biomaterials such as collagen [55] or silk [56][57][58][59] cause a significantly lower immune response. Still, synthetic polymers remain attractive substrates for tissue engineering and can be functionalized to enhance their biocompatibility. For example, magnesium hydroxide nanoparticles may help to neutralize pH changes by PLGA degradation acidic products, alleviating inflammation [60].
Hydrophilicity or hydrophobicity of the scaffold is another important characteristic affecting the protein adsorption and therefore the host immune response. Most synthetic polymers are hydrophobic, which correlates with high immunogenicity, but hydrophilic molecules such as polyethylene oxide (PEO) [61], polyethylene glycol (PEG) [61], or graphene oxide (GO) [62] can be used to modify their surface chemistry [63]. There is evidence that compound scaffolds consisting of both synthetic and natural biomaterials show rather good biocompatibility, for example, collagen from micronized porcine cartilage alleviated the inflammatory effect of a PLGA scaffold in a rat model [54]. However, the best available option in terms of biocompatibility remains decellularized ECM (dECM). Not only does dECM provide a perfect microenvironment for cells, it was also repeatedly reported to have immunomodulatory properties, including the influence on the macrophages, namely their polarization to the anti-inflammatory M2 phenotype [63][64][65][66]. Unsurprisingly, many researchers try to mimic ECM properties when designing biomaterials [67].
Some researchers propose scaffold-free approaches to avoid any possible adverse immune reactions to the biomaterials. Both chondrocyte-based [68] and synovial mesenchymal stem cells (MSCs)-based [69] cell sheets were demonstrated to promote good cartilage regeneration without any undesired inflammatory response in vivo. However, although considered “scaffold-free”, both constructs contained the ECM synthesized by the cells [70], and therefore these data may speak in favor of the use of ECM as well.

4.2. Cells

MSCs from different sources, the cell type the most extensively used in tissue engineering, are appreciated for their low immunogenicity and certain immunosuppressive capacity [71]. However, it was reported that in the process of differentiation in vivo their immunogenicity is induced due to MHC-I and MHC-II expression [72], and there is evidence that MSCs can cause a memory T-cell response in immunocompetent hosts [73].
MSCs’ therapeutic potential has long been discussed in the context of their differentiation capacity, i.e., ability to replace damaged tissues. However, a growing body of evidence suggests that MSCs exert their regenerative effect via paracrine activity [74]. Cytokines, chemokines, and, most importantly, extracellular vesicles (EVs) secreted by MSCs are now considered the key players in MSC-based therapies. EVs are heterogeneous membrane nanoparticles providing intercellular communication. Their research and use in various fields of regenerative medicine have been boosted over the past years, and they were demonstrated to possess all the advantages of MSCs without considerable drawbacks. For example, MSC-derived EVs exhibit no risk of tumor formation and demonstrate even lower immunogenicity than MSCs [75]. Moreover, they have lower storage demands and are therefore a promising agent for biomedical product development. Summing up, this makes cell-free approaches more and more prevalent in tissue engineering, including cartilage repair [76].

4.3. Biochemical Factors

Cytokines and growth factors control inflammation as well as cell proliferation, migration, and differentiation, thus orchestrating tissue remodeling and regeneration. Biochemical factors are naturally synthesized by the cellular component of tissue-engineered constructs: for example, MSCs are known to produce a variety of both growth factors and cytokines [77][78]. Moreover, cell cultures can be transfected or transduced so that the cells will secrete the desired bioactive molecules. In a study conducted by Holladay and colleagues, transfected rat bone-marrow-derived MSCs over-expressing an anti-inflammatory cytokine interleukin 10 (IL-10) demonstrated a good retention rate within collagen scaffolds in vivo for up to 7 days in comparison to unmodified cells, while the number of inflammatory cells during this period was decreased [79]. However, the authors reported that IL-10 modified the MSCs’ retention rate to reduce almost to the same level as unmodified cells by day 21, with an unexpected increase in inflammatory cell number on day 7, speculating that prolonged culturing might have altered the MSCs’ phenotype to become more immunogenic. Thus, given the difficulty to control implanted cells in vivo as well as the growing popularity of cell-free approaches, scaffold loading with bioactive factors has been developed. For example, in the same study, Holladay and colleagues proposed an alternative method where rat bone-marrow-derived MSCs were implanted in scaffolds loaded with IL-10 plasmid–polymer complexes, or “polyplexes”, allowing in vivo transfection [79]. This approach increased the MSCs’ retention rates for up to 21 days, which led to prolonged IL-10 release and decreased number of inflammatory cells. Although the idea of scaffold loading with bioactive molecules is not a new one [80], their delivery remains a challenge and ranges from chemical [81][82] or physical [83] incorporation to the use of micro- [84][85][86] or nanoparticles [87]. In this light, ECM as a natural source of bioactive molecules, such as VEGF [88], granulocyte-macrophage colony-stimulating factor (G-MCSF) [66], hepatocyte growth factor (HGF) [66][88], transforming growth factor-beta (TGFβ) [66][88], interleukin 3 (IL-3) [66], or interferon-gamma (INFγ) [66], is of great interest. Moreover, while state-of-the-art approaches suggest EVs’ integration into scaffolds as a powerful tool for cell signaling [89][90][91], ECM-based scaffolds are a natural source of matrix-bound nanovesicles (MBVs), a subgroup of EVs known for their unique immunomodulatory properties [92].

References

  1. Oleksyszyn, J.; Augustine, A.J. Plasminogen modulation of IL-1-stimulated degradation in bovine and human articular cartilage explants. The role of the endogenous inhibitors: PAI-1,α 2-antiplasmin,α 1-PI,α 2-macroglobulin and TIMP. Agents Actions 1996, 45, 464–472.
  2. Pascual, E.; Jovaní, V. Synovial fluid analysis. Best Pract. Res. Clin. Rheumatol. 2005, 19, 371–386.
  3. Gerwin, N.; Hops, C.; Lucke, A. Intraarticular drug delivery in osteoarthritis. Adv. Drug Deliv. Rev. 2006, 58, 226–242.
  4. Brown, J.M.; Mantoku, A.; Sabokbar, A.; Oppermann, U.; Hassan, A.B.; Kudo, A.; Athanasou, N. Periostin expression in neoplastic and non-neoplastic diseases of bone and joint. Clin. Sarcoma Res. 2018, 8, 1–8.
  5. Zhou, L.; Gu, M.; Ma, X.; Wen, L.; Zhang, B.; Lin, Y.; Pan, J. Long non-coding RNA PCAT-1 regulates apoptosis of chondrocytes in osteoarthritis by sponging miR-27b-3p. J. Bone Miner. Metab. 2021, 39, 139–147.
  6. Bosch, M.H.V.D. Inflammation in osteoarthritis: Is it time to dampen the alarm(in) in this debilitating disease? Clin. Exp. Immunol. 2019, 195, 153–166.
  7. Sokolove, J.; Lepus, C.M. Role of inflammation in the pathogenesis of osteoarthritis: Latest findings and interpretations. Ther. Adv. Musculoskelet. Dis. 2013, 5, 77–94.
  8. Peshkova, M.; Lychagin, A.; Lipina, M.; Di Matteo, B.; Anzillotti, G.; Ronzoni, F.; Kosheleva, N.; Shpichka, A.; Royuk, V.; Fomin, V.; et al. Gender-Related Aspects in Osteoarthritis Development and Progression: A Review. Int. J. Mol. Sci. 2022, 23, 2767.
  9. Ayral, X.; Pickering, E.; Woodworth, T.; 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.
  10. Krasnokutsky, S.; Belitskaya-Lévy, I.; Bencardino, J.; Samuels, J.; Attur, M.; Regatte, R.; Rosenthal, P.; Greenberg, J.; Schweitzer, M.; Abramson, S.B.; et al. Quantitative magnetic resonance imaging evidence of synovial proliferation is associated with radiographic severity of knee osteoarthritis. Arthritis Rheum. 2011, 63, 2983–2991.
  11. Bannuru, R.R.; Osani, M.C.; Vaysbrot, E.E.; Arden, N.K.; Bennell, K.; Bierma-Zeinstra, S.M.A.; Kraus, V.B.; Lohmander, L.S.; Abbott, J.H.; Bhandari, M.; et al. OARSI guidelines for the non-surgical management of knee, hip, and polyarticular osteoarthritis. Osteoarthr. Cartil. 2019, 27, 1578–1589.
  12. Cao, P.; Li, Y.; Tang, Y.; Ding, C.; Hunter, D.J. Pharmacotherapy for knee osteoarthritis: Current and emerging therapies. Expert Opin. Pharmacother. 2020, 21, 797–809.
  13. Robinson, P.D.; McEwan, J.; Adukia, V.; Prabhakar, M. Osteoarthritis and arthroplasty of the hip and knee. Br. J. Hosp. Med. 2018, 79, C54–C59.
  14. Xie, C.; Chen, Q. Adipokines: New Therapeutic Target for Osteoarthritis? Curr. Rheumatol. Rep. 2019, 21, 71.
  15. Tu, C.; He, J.; Wu, B.; Wang, W.; Li, Z. An extensive review regarding the adipokines in the pathogenesis and progression of osteoarthritis. Cytokine 2019, 113, 1–12.
  16. Makris, E.A.; Gomoll, A.H.; Malizos, K.N.; Hu, J.C.; Athanasiou, K.A. Repair and tissue engineering techniques for articular cartilage. Nat. Rev. Rheumatol. 2015, 11, 21–34.
  17. Wang, Z.; Le, H.; Wang, Y.; Liu, H.; Li, Z.; Yang, X.; Wang, C.; Ding, J.; Chen, X. Instructive cartilage regeneration modalities with advanced therapeutic implantations under abnormal conditions. Bioact. Mater. 2021, 11, 317–338.
  18. Iwanaga, T.; Shikichi, M.; Kitamura, H.; Yanase, H.; Nozawa-Inoue, K. Morphology and Functional Roles of Synoviocytes in the Joint. Arch. Histol. Cytol. 2000, 63, 17–31.
  19. Robinson, W.H.; Lepus, C.M.; Wang, Q.; Raghu, H.; Mao, R.; Lindstrom, T.M.; Sokolove, J. Low-grade inflammation as a key mediator of the pathogenesis of osteoarthritis. Nat. Rev. Rheumatol. 2016, 12, 580–592.
  20. Pazár, B.; Ea, H.-K.; Narayan, S.; Kolly, L.; Bagnoud, N.; Chobaz, V.; Roger, T.; Lioté, F.; So, A.; Busso, N. Basic Calcium Phosphate Crystals Induce Monocyte/Macrophage IL-1β Secretion through the NLRP3 Inflammasome In Vitro. J. Immunol. 2011, 186, 2495–2502.
  21. Denoble, A.E.; Huffman, K.M.; Stabler, T.V.; Kelly, S.J.; Hershfield, M.S.; McDaniel, G.E.; Coleman, R.E.; Kraus, V.B. Uric acid is a danger signal of increasing risk for osteoarthritis through inflammasome activation. Proc. Natl. Acad. Sci. USA 2011, 108, 2088–2093.
  22. Sillat, T.; Barreto, G.; Clarijs, P.; Soininen, A.; Ainola, M.; Pajarinen, J.; Korhonen, M.; Konttinen, Y.T.; Sakalyte, R.; Hukkanen, M.; et al. Toll-like receptors in human chondrocytes and osteoarthritic cartilage. Acta Orthop. 2013, 84, 585–592.
  23. Kim, H.A.; Cho, M.-L.; Choi, H.Y.; Yoon, C.S.; Jhun, J.Y.; Oh, H.J.; Kim, H.-Y. The catabolic pathway mediated by Toll-like receptors in human osteoarthritic chondrocytes. Arthritis Rheum. 2006, 54, 2152–2163.
  24. Berenbaum, F. Osteoarthritis as an inflammatory disease (osteoarthritis is not osteoarthrosis!). Osteoarthr. Cartil. 2013, 21, 16–21.
  25. Kennedy, A.; Fearon, U.; Veale, D.J.; Godson, C. Macrophages in Synovial Inflammation. Front. Immunol. 2011, 2, 52.
  26. Fernandes, T.L.; Gomoll, A.H.; Lattermann, C.; Hernandez, A.J.; Bueno, D.F.; Amano, M.T. Macrophage: A Potential Target on Cartilage Regeneration. Front. Immunol. 2020, 11, 111.
  27. de Lange-Brokaar, B.; Ioan-Facsinay, A.; van Osch, G.; Zuurmond, A.-M.; Schoones, J.; Toes, R.; Huizinga, T.; Kloppenburg, M. Synovial inflammation, immune cells and their cytokines in osteoarthritis: A review. Osteoarthr. Cartil. 2012, 20, 1484–1499.
  28. Vincent, H.K.; Percival, S.S.; Conrad, B.P.; Seay, A.N.; Montero, C.; Vincent, K.R. Hyaluronic Acid (HA) Viscosupplementation on Synovial Fluid Inflammation in Knee Osteoarthritis: A Pilot Study. Open Orthop. J. 2013, 7, 378–384.
  29. Van Lent, P.L.E.M.; Blom, A.B.; Van Der Kraan, P.; Holthuysen, A.E.M.; Vitters, E.; Van Rooijen, N.; Smeets, R.L.; Nabbe, K.C.A.M.; Berg, W.B.V.D. Crucial role of synovial lining macrophages in the promotion of transforming growth factor?-mediated osteophyte formation. Arthritis Rheum. 2004, 50, 103–111.
  30. Blom, A.B.; van Lent, P.L.; Holthuysen, A.E.; van der Kraan, P.M.; Roth, J.; van Rooijen, N.; Berg, W.B.V.D. Synovial lining macrophages mediate osteophyte formation during experimental osteoarthritis. Osteoarthr. Cartil. 2004, 12, 627–635.
  31. Blom, A.B.; van Lent, P.L.; Libregts, S.; Holthuysen, A.E.; van der Kraan, P.M.; van Rooijen, N.; Berg, W.B.V.D. Crucial role of macrophages in matrix metalloproteinase–mediated cartilage destruction during experimental osteoarthritis: Involvement of matrix metalloproteinase 3. Arthritis Rheum. 2007, 56, 147–157.
  32. Wu, C.-L.; McNeill, J.; Goon, K.; Little, D.; Kimmerling, K.; Huebner, J.; Kraus, V.; Guilak, F. Conditional Macrophage Depletion Increases Inflammation and Does Not Inhibit the Development of Osteoarthritis in Obese Macrophage Fas-Induced Apoptosis-Transgenic Mice. Arthritis Rheumatol. 2017, 69, 1772–1783.
  33. Wright, H.; Moots, R.J.; Edwards, S.W. The multifactorial role of neutrophils in rheumatoid arthritis. Nat. Rev. Rheumatol. 2014, 10, 593–601.
  34. Punzi, L.; Galozzi, P.; Luisetto, R.; Favero, M.; Ramonda, R.; Oliviero, F.; Scanu, A. Post-traumatic arthritis: Overview on pathogenic mechanisms and role of inflammation. RMD Open 2016, 2, e000279.
  35. Sostres, C.; Gargallo, C.J.; Lanas, A. Nonsteroidal anti-inflammatory drugs and upper and lower gastrointestinal mucosal damage. Arthritis Res. Ther. 2013, 15 (Suppl. S3), S3–S8.
  36. Bally, M.; Dendukuri, N.; Rich, B.; Nadeau, L.; Helin-Salmivaara, A.; Garbe, E.; Brophy, J. Risk of acute myocardial infarction with NSAIDs in real world use: Bayesian meta-analysis of individual patient data. BMJ 2017, 357, j1909.
  37. Popiolek, I.; Hydzik, P.; Jagielski, P.; Zrodlowska, M.; Mystek, K.; Porebski, G. Risk Factors for Hepatotoxicity Due to Paracetamol Overdose in Adults. Medicina 2021, 57, 752.
  38. Roberts, E.; Nunes, V.D.; Buckner, S.; Latchem, S.; Constanti, M.; Miller, P.; Doherty, M.; Zhang, W.; Birrell, F.; Porcheret, M.; et al. Paracetamol: Not as safe as we thought? A systematic literature review of observational studies. Ann. Rheum. Dis. 2016, 75, 552–559.
  39. Kraus, V.; Birmingham, J.; Stabler, T.; Feng, S.; Taylor, D.; Moorman, C.; Garrett, W.; Toth, A. Effects of intraarticular IL1-Ra for acute anterior cruciate ligament knee injury: A randomized controlled pilot trial (NCT00332254). Osteoarthr. Cartil. 2012, 20, 271–278.
  40. Chevalier, X.; Goupille, P.; Beaulieu, A.D.; Burch, F.X.; Bensen, W.G.; Conrozier, T.; Loeuille, D.; Kivitz, A.J.; Silver, D.; Appleton, B.E. Intraarticular injection of anakinra in osteoarthritis of the knee: A multicenter, randomized, double-blind, placebo-controlled study. Arthritis Rheum. 2009, 61, 344–352.
  41. Cohen, S.B.; Proudman, S.; Kivitz, A.J.; Burch, F.X.; Donohue, J.P.; Burstein, D.; Sun, Y.-N.; Banfield, C.; Vincent, M.S.; Ni, L.; et al. A randomized, double-blind study of AMG 108 (a fully human monoclonal antibody to IL-1R1) in patients with osteoarthritis of the knee. Arthritis Res. Ther. 2011, 13, R125.
  42. Verbruggen, A.; Wittoek, R.; Cruyssen, B.V.; Elewaut, D. Tumour necrosis factor blockade for the treatment of erosive osteoarthritis of the interphalangeal finger joints: A double blind, randomised trial on structure modification. Ann. Rheum. Dis. 2012, 71, 891–898.
  43. Maksymowych, W.P.; Russell, A.S.; Chiu, P.; Yan, A.; Jones, N.; Clare, T.; Lambert, R.G. Targeting tumour necrosis factor alleviates signs and symptoms of inflammatory osteoarthritis of the knee. Arthritis Res. Ther. 2012, 14, R206.
  44. Fioravanti, A.; Fabbroni, M.; Cerase, A.; Galeazzi, M. Treatment of erosive osteoarthritis of the hands by intra-articular infliximab injections: A pilot study. Rheumatol. Int. 2009, 29, 961–965.
  45. Chevalier, X.; Ravaud, P.; Maheu, E.; Baron, G.; Rialland, A.; Vergnaud, P.; Roux, C.; Maugars, Y.; Mulleman, D.; Lukas, C.; et al. Adalimumab in patients with hand osteoarthritis refractory to analgesics and NSAIDs: A randomised, multicentre, double-blind, placebo-controlled trial. Ann. Rheum. Dis. 2015, 74, 1697–1705.
  46. Avouac, J.; Vicaut, E.; Bardin, T.; Richette, P. Efficacy of joint lavage in knee osteoarthritis: Meta-analysis of randomized controlled studies. Rheumatology 2010, 49, 334–340.
  47. Gao, L.; Goebel, L.K.H.; Orth, P.; Cucchiarini, M.; Madry, H. Subchondral drilling for articular cartilage repair: A systematic review of translational research. Dis. Model. Mech. 2018, 11, dmm034280.
  48. Schreiner, M.M.; Raudner, M.; Szomolanyi, P.; Ohel, K.; Ben-Zur, L.; Juras, V.; Mlynarik, V.; Windhager, R.; Trattnig, S. Chondral and Osteochondral Femoral Cartilage Lesions Treated with GelrinC: Significant Improvement of Radiological Outcome Over Time and Zonal Variation of the Repair Tissue Based on T2 Mapping at 24 Months. Cartilage 2021, 13 (Suppl. S1), 604S–616S.
  49. Murphy, M.P.; Koepke, L.S.; Lopez, M.T.; Tong, X.; Ambrosi, T.H.; Gulati, G.S.; Marecic, O.; Wang, Y.; Ransom, R.C.; Hoover, M.Y.; et al. Articular cartilage regeneration by activated skeletal stem cells. Nat. Med. 2020, 26, 1583–1592.
  50. Chimutengwende-Gordon, M.; Donaldson, J.; Bentley, G. Current solutions for the treatment of chronic articular cartilage defects in the knee. EFORT Open Rev. 2020, 5, 156–163.
  51. Hevesi, M.; Jacob, G.; Shimomura, K.; Ando, W.; Nakamura, N.; Krych, A.J. Current hip cartilage regeneration/repair modalities: A scoping review of biologics and surgery. Int. Orthop. 2021, 45, 319–333.
  52. Yang, F.; Zhang, Y.; Liu, B.; Cao, M.; Yang, J.; Tian, F.; Yang, P.; Qin, K.; Zhao, D. Basic fibroblast growth factor and agarose gel promote the ability of immune privilege of allogeneic cartilage transplantation in rats. J. Orthop. Transl. 2019, 22, 73–80.
  53. Asawa, Y.; Sakamoto, T.; Komura, M.; Watanabe, M.; Nishizawa, S.; Takazawa, Y.; Takato, T.; Hoshi, K. Early Stage Foreign Body Reaction against Biodegradable Polymer Scaffolds Affects Tissue Regeneration during the Autologous Transplantation of Tissue-Engineered Cartilage in the Canine Model. Cell Transplant. 2012, 21, 1431–1442.
  54. Kim, S.; Jang, J.E.; Lee, J.H.; Khang, G. Composite scaffold of micronized porcine cartilage/poly(lactic-co-glycolic acid) enhances anti-inflammatory effect. Mater. Sci. Eng. C 2018, 88, 46–52.
  55. van Putten, S.M.; Ploeger, D.T.; Popa, E.R.; Bank, R.A. Macrophage phenotypes in the collagen-induced foreign body reaction in rats. Acta Biomater. 2013, 9, 6502–6510.
  56. Meinel, L.; Hofmann, S.; Karageorgiou, V.; Kirker-Head, C.; McCool, J.; Gronowicz, G.; Zichner, L.; Langer, R.; Vunjak-Novakovic, G.; Kaplan, D.L. The inflammatory responses to silk films in vitro and in vivo. Biomaterials 2005, 26, 147–155.
  57. Aramwit, P.; Kanokpanont, S.; De-Eknamkul, W.; Srichana, T. Monitoring of inflammatory mediators induced by silk sericin. J. Biosci. Bioeng. 2009, 107, 556–561.
  58. Zhang, Y.; Wu, C.; Luo, T.; Li, S.; Cheng, X.; Miron, R.J. Synthesis and inflammatory response of a novel silk fibroin scaffold containing BMP7 adenovirus for bone regeneration. Bone 2012, 51, 704–713.
  59. Qi, C.; Liu, J.; Jin, Y.; Xu, L.; Wang, G.; Wang, Z.; Wang, L. Photo-crosslinkable, injectable sericin hydrogel as 3D biomimetic extracellular matrix for minimally invasive repairing cartilage. Biomaterials 2018, 163, 89–104.
  60. Park, K.-S.; Kim, B.-J.; Lih, E.; Park, W.; Lee, S.-H.; Joung, Y.K.; Han, D.K. Versatile effects of magnesium hydroxide nanoparticles in PLGA scaffold–mediated chondrogenesis. Acta Biomater. 2018, 73, 204–216.
  61. Drury, J.L.; Mooney, D.J. Hydrogels for tissue engineering: Scaffold design variables and applications. Biomaterials 2003, 24, 4337–4351.
  62. Aidun, A.; Firoozabady, A.S.; Moharrami, M.; Ahmadi, A.; Haghighipour, N.; Bonakdar, S.; Faghihi, S. Graphene oxide incorporated polycaprolactone/chitosan/collagen electrospun scaffold: Enhanced osteogenic properties for bone tissue engineering. Artif. Organs 2019, 43, E264–E281.
  63. Wei, F.; Liu, S.; Chen, M.; Tian, G.; Zha, K.; Yang, Z.; Jiang, S.; Li, M.; Sui, X.; Chen, Z.; et al. Host Response to Biomaterials for Cartilage Tissue Engineering: Key to Remodeling. Front. Bioeng. Biotechnol. 2021, 9, 664592.
  64. Fishman, J.M.; Lowdell, M.W.; Urbani, L.; Ansari, T.; Burns, A.J.; Turmaine, M.; North, J.; Sibbons, P.; Seifalian, A.M.; Wood, K.J.; et al. Immunomodulatory effect of a decellularized skeletal muscle scaffold in a discordant xenotransplantation model. Proc. Natl. Acad. Sci. USA 2013, 110, 14360–14365.
  65. He, C.; Yang, Z.; Jin, Y.; Qi, X.; Chu, J.; Deng, X. ADM Scaffolds Generate a Pro-regenerative Microenvironment During Full-Thickness Cutaneous Wound Healing Through M2 Macrophage Polarization via Lamtor1. Front. Physiol. 2018, 9, 657.
  66. Londono, R.; Badylak, S.F. Biologic Scaffolds for Regenerative Medicine: Mechanisms of In vivo Remodeling. Ann. Biomed. Eng. 2015, 43, 577–592.
  67. He, W.; Bai, J.; Chen, X.; Suo, D.; Wang, S.; Guo, Q.; Yin, W.; Geng, D.; Wang, M.; Pan, G.; et al. Reversible dougong structured receptor–ligand recognition for building dynamic extracellular matrix mimics. Proc. Natl. Acad. Sci. USA 2022, 119, e2117221119.
  68. Wang, F.; Hu, Y.; He, D.; Zhou, G.; Ellis, E. Scaffold-free cartilage cell sheet combined with bone-phase BMSCs-scaffold regenerate osteochondral construct in mini-pig model. Am. J. Transl. Res. 2018, 10, 2997–3010.
  69. Koizumi, K.; Ebina, K.; Hart, D.A.; Hirao, M.; Noguchi, T.; Sugita, N.; Yasui, Y.; Chijimatsu, R.; Yoshikawa, H.; Nakamura, N. Synovial mesenchymal stem cells from osteo- or rheumatoid arthritis joints exhibit good potential for cartilage repair using a scaffold-free tissue engineering approach. Osteoarthr. Cartil. 2016, 24, 1413–1422.
  70. Efremov, Y.M.; Zurina, I.M.; Presniakova, V.S.; Kosheleva, N.V.; Butnaru, D.V.; Svistunov, A.A.; Rochev, Y.A.; Timashev, P.S. Mechanical properties of cell sheets and spheroids: The link between single cells and complex tissues. Biophys. Rev. 2021, 13, 541–561.
  71. Gu, L.-H.; Zhang, T.-T.; Li, Y.; Yan, H.-J.; Qi, H.; Li, F.-R. Immunogenicity of allogeneic mesenchymal stem cells transplanted via different routes in diabetic rats. Cell. Mol. Immunol. 2014, 12, 444–455.
  72. Huang, X.-P.; Sun, Z.; Miyagi, Y.; Kinkaid, H.M.; Zhang, L.; Weisel, R.D.; Li, R.-K. Differentiation of Allogeneic Mesenchymal Stem Cells Induces Immunogenicity and Limits Their Long-Term Benefits for Myocardial Repair. Circulation 2010, 122, 2419–2429.
  73. Nauta, A.J.; Westerhuis, G.; Kruisselbrink, A.B.; Lurvink, E.G.A.; Willemze, R.; Fibbe, W.E. Donor-derived mesenchymal stem cells are immunogenic in an allogeneic host and stimulate donor graft rejection in a nonmyeloablative setting. Blood 2006, 108, 2114–2120.
  74. Kusuma, G.; Li, A.; Zhu, D.; McDonald, H.; Chambers, D.; Frith, J.; Lim, R. Engineering mesenchymal stem cell paracrine activity with 3D culture. Cytotherapy 2020, 22, S51.
  75. Yin, K.; Wang, S.; Zhao, R.C. Exosomes from mesenchymal stem/stromal cells: A new therapeutic paradigm. Biomark. Res. 2019, 7, 1–8.
  76. Chen, P.; Zheng, L.; Wang, Y.; Tao, M.; Xie, Z.; Xia, C.; Gu, C.; Chen, J.; Qiu, P.; Mei, S.; et al. Desktop-stereolithography 3D printing of a radially oriented extracellular matrix/mesenchymal stem cell exosome bioink for osteochondral defect regeneration. Theranostics 2019, 9, 2439–2459.
  77. Leuning, D.G.; Beijer, N.; du Fossé, N.; Vermeulen, S.; Lievers, E.; Van Kooten, C.; Rabelink, T.; De Boer, J. The cytokine secretion profile of mesenchymal stromal cells is determined by surface structure of the microenvironment. Sci. Rep. 2018, 8, 7716.
  78. Eom, Y.W.; Oh, J.-E.; Lee, J.I.; Baik, S.K.; Rhee, K.-J.; Shin, H.C.; Kim, Y.M.; Ahn, C.M.; Kong, J.H.; Kim, H.S.; et al. The role of growth factors in maintenance of stemness in bone marrow-derived mesenchymal stem cells. Biochem. Biophys. Res. Commun. 2014, 445, 16–22.
  79. Holladay, C.; Power, K.; Sefton, M.; O’Brien, T.; Gallagher, W.; Pandit, A. Functionalized Scaffold-mediated Interleukin 10 Gene Delivery Significantly Improves Survival Rates of Stem Cells In Vivo. Mol. Ther. 2011, 19, 969–978.
  80. Lee, S.J. Cytokine delivery and tissue engineering. Yonsei Med. J. 2000, 41, 704–719.
  81. Sridhar, B.V.; Doyle, N.R.; Randolph, M.A.; Anseth, K.S. Covalently tethered TGF- β 1 with encapsulated chondrocytes in a PEG hydrogel system enhances extracellular matrix production. J. Biomed. Mater. Res. Part A 2014, 102, 4464–4472.
  82. Cavalli, E.; Levinson, C.; Hertl, M.; Broguiere, N.; Brück, O.; Mustjoki, S.; Gerstenberg, A.; Weber, D.; Salzmann, G.; Steinwachs, M.; et al. Characterization of polydactyly chondrocytes and their use in cartilage engineering. Sci. Rep. 2019, 9, 4275.
  83. Zhou, T.; Li, X.; Li, G.; Tian, T.; Lin, S.; Shi, S.; Liao, J.; Cai, X.; Lin, Y. Injectable and thermosensitive TGF-β1-loaded PCEC hydrogel system for in vivo cartilage repair. Sci. Rep. 2017, 7, 10553.
  84. Bian, L.; Zhai, D.Y.; Tous, E.; Rai, R.; Mauck, R.L.; Burdick, J.A. Enhanced MSC chondrogenesis following delivery of TGF-β3 from alginate microspheres within hyaluronic acid hydrogels in vitro and in vivo. Biomaterials 2011, 32, 6425–6434.
  85. Guo, X.; Park, H.; Young, S.; Kretlow, J.D.; Beucken, J.J.V.D.; Baggett, L.S.; Tabata, Y.; Kasper, F.K.; Mikos, A.G.; Jansen, J.A. Repair of osteochondral defects with biodegradable hydrogel composites encapsulating marrow mesenchymal stem cells in a rabbit model. Acta Biomater. 2010, 6, 39–47.
  86. Elisseeff, J.; McIntosh, W.; Fu, K.; Blunk, T.; Langer, R. Controlled-release of IGF-I and TGF-β1 in a photopolymerizing hydrogel for cartilage tissue engineering. J. Orthop. Res. 2001, 19, 1098–1104.
  87. Wei, P.; Xu, Y.; Gu, Y.; Yao, Q.; Li, J.; Wang, L. IGF-1-releasing PLGA nanoparticles modified 3D printed PCL scaffolds for cartilage tissue engineering. Drug Deliv. 2020, 27, 1106–1114.
  88. Boyd, D.F.; Thomas, P.G. Towards integrating extracellular matrix and immunological pathways. Cytokine 2017, 98, 79–86.
  89. Yan, H.-C.; Yu, T.-T.; Li, J.; Qiao, Y.-Q.; Wang, L.-C.; Zhang, T.; Li, Q.; Zhou, Y.-H.; Liu, D.-W. The Delivery of Extracellular Vesicles Loaded in Biomaterial Scaffolds for Bone Regeneration. Front. Bioeng. Biotechnol. 2020, 8, 1015.
  90. Kang, M.; Lee, C.-S.; Lee, M. Bioactive Scaffolds Integrated with Liposomal or Extracellular Vesicles for Bone Regeneration. Bioengineering 2021, 8, 137.
  91. Liu, Y.; Ma, Y.; Zhang, J.; Yuan, Y.; Wang, J. Exosomes: A Novel Therapeutic Agent for Cartilage and Bone Tissue Regeneration. Dose-Response 2019, 17, 1559325819892702.
  92. Huleihel, L.; Hussey, G.S.; Naranjo, J.D.; Zhang, L.; Dziki, J.L.; Turner, N.J.; Stolz, D.B.; Badylak, S.F. Matrix-bound nanovesicles within ECM bioscaffolds. Sci. Adv. 2016, 2, e1600502.
More
Information
Subjects: Others
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , , , , , ,
View Times: 300
Revisions: 2 times (View History)
Update Date: 22 Nov 2022
1000/1000