Optimizing Tenogenic Differentiation of Equine Adipose-Derived Mesenchymal Stem Cells (eq-ASC) Using TGFB3 Along with BMP Antagonists

Document Type : Original Article

Authors

1 Division of Physiology, Department of Basic Sciences, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran

2 Department of Animal Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran

3 Stem Cell Biology and Regenerative Medicine Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran

Abstract

Objective: Tendon repair strategies usually are accompanied by pathological mineralization and scar tissue formation that increases the risk of re-injuries. This study aimed to establish an efficient tendon regeneration method simultaneously with a reduced risk of ectopic bone formation.
Materials and Methods: In this experimental study, tenogenic differentiation was induced through transforming growth factor- β3 (TGFB3) treatment in combination with the inhibiting concentrations of bone morphogenetic proteins (BMP) antagonists, gremlin-2 (GREM2), and a Wnt inhibitor, namely sclerostin (SOST). The procedure’s efficacy was evaluated using real-time polymerase chain reaction (qPCR) for expression analysis of tenogenic markers and osteochondrogenic marker genes. The expression level of two tenogenic markers, SCX and MKX, was also evaluated by immunocytochemistry. Sirius Red staining was performed to examine the amounts of collagen fibers. Moreover, to investigate the impact of the substrate on tenogenic differentiation, the nanofibrous scaffolds that highly resemble tendon extracellular matrix was employed.
Results: Aggregated features formed in spontaneous normal culture conditions followed by up-regulation of tenogenic
and osteogenic marker genes, including SCX, MKX, COL1A1, RUNX2, and CTNNB1. TGFB3 treatment exaggerated
morphological changes and markedly amplified tenogenic differentiation in a shorter period of time. Along with TGFB3 treatment, inhibition of BMPs by GREM2 and SOST delayed migratory events to some extent and dramatically reduced osteo-chondrogenic markers synergistically. Nanofibrous scaffolds increased tenogenic markers while declining the expression of osteo-chondrogenic genes.
Conclusion: These findings revealed an appropriate in vitro potential of spontaneous tenogenic differentiation of eq- ASCs that can be improved by simultaneous activation of TGFB and inhibition of osteoinductive signaling pathways.

Keywords


1. Ortved KF. Regenerative Medicine and rehabilitation for tendinous and ligamentous injuries in sport horses. Vet Clin North Am Equine Pract. 2018; 34(2): 359-373.
2. Yang G, Rothrauff BB,Tuan RS. Tendon and ligament regeneration and repair: clinical relevance and developmental paradigm. Birth Defects Res C Embryo Today. 2013; 99(3): 203-222.
3. Conrad S, Weber K, Walliser U, Geburek F, Skutella T. Stem cell therapy for tendon regeneration: current status and future directionsSeries. Adv Exp Med Biol. 2019; 1084: 61-93.
4. Becerra P, Valdes Vazquez MA, Dudhia J, Fiske-Jackson AR, Neves F, Hartman NG, et al. Distribution of injected technetium (99m)-labeled mesenchymal stem cells in horses with naturally occurring tendinopathy. J Orthop Res. 2013; 31(7): 1096-1102.
5. Burk J, Gittel C, Heller S, Pfeiffer B, Paebst F, Ahrberg AB, et al. Gene expression of tendon markers in mesenchymal stromal cells derived from different sources. BMC Res Notes. 2014; 7(1): 826.
6. Molloy T, Wang Y, Murrell G. The roles of growth factors in tendon and ligament healing. Sports Med. 2003; 33(5): 381-394.
7. Mishra A, Woodall Jr, Vieira A. Treatment of tendon and muscle using platelet-rich plasma. Clin Sports Med. 2009; 28(1): 113-125.
8. Nourissat G, Berenbaum F, Duprez D. Tendon injury: from biology to tendon repair. Nat Rev Rheumatol. 2015; 11(4): 223-233.
9. Wang MK, Sun HQ, Xiang YC, Jiang F, Su YP, Zou ZM. Different roles of TGF- β in the multi-lineage differentiation of stem cells. World J of Stem Cells. 2012; 4(5): 28-34.
10. Chien C, Pryce B, Tufa SF, Keene DR, Huang AH. Optimizing a 3D model system for molecular manipulation of tenogenesis. Connect Tissue Res. 2018; 59(4): 295-308.
11. Docheva D, Müller SA, Majewski M, Evans CH. Biologics for tendon repair. Adv Drug Deliv Rev. 2015; 84: 222-239.
12. Kaji DA, Howell KL, Balic Z, Hubmacher D, Huang AH. Tgfβ signaling is required for tenocyte recruitment and functional neonatal tendon regeneration. Elife. 2020; 9: e51779.
13. Hou Y, Mao Z, Wei X, Lin L, Chen L, Wang H, et al. The roles of TGF-beta1 gene transfer on collagen formation during achilles tendon healing. Biochem Biophys Res Commun. 2009; 383(2): 235-239.
14. Campbell BH, Agarwal C, Wang JH. TGF-beta1, TGF-beta3, and PGE(2) regulate contraction of human patellar tendon fibroblasts. Biomech Model Mechanobiol. 2004; 2(4): 239-245.
15. Grafe I, Alexander S, Peterson JR, Snider TN, Levi B, Lee B, et al. TGF-β family signaling in mesenchymal differentiation. Cold Spring Harb Perspect Biol. 2018; 10(5): a022202.
16. Cao Y, LV Q, LV C. MicroRNA-153 suppresses the osteogenic differentiation of human mesenchymal stem cells by targeting bone morphogenetic protein receptor type II. Int J Mol Med. 2015; 36(3): 760-766.
17. Shen H, Gelberman RH, Silva MJ, Sakiyama-Elbert SE, Thomopoulos S. BMP12 induces tenogenic differentiation of adiposederived stromal cells. PLoS One. 2013; 8(10): e77613.
18. Chen X, Yin Z, Chen JL, Shen WL, Liu HH, Tang QM, et al. Force and scleraxis synergistically promote the commitment of human ES cells derived MSCs to tenocytes. Sci Rep. 2012; 2: 977.
19. Chang C. Agonists and antagonists of TGF-β family ligands. Cold Spring Harb Perspect Biol. 2016; 8(8): a021923.
20. Semënov M, Tamai K, He X. SOST is a ligand for LRP5/LRP6 and a Wnt signaling inhibitor. J Biol Chem. 2005; 280(29): 26770-26775.
21. Lui PP. Histopathological changes in tendinopathy-potential roles of BMPs? Rheumatology (Oxford). 2013; 52(12): 2116-2126.
22. Zhang X, Bogdanowicz D, Erisken C, Lee NM, Lu HH. Biomimetic scaffold design for functional and integrative tendon repair. J Shoulder Elbow Surg. 2012; 21(2): 266-277.
23. Webb WR, Dale TP, Lomas AJ, Zeng G, Wimpenny I, El Haj AJ, et al. The application of poly(3-hydroxybutyrate-co-3-hydroxyhex anoate) scaffolds for tendon repair in the rat model. Biomaterials. 2013; 34(28): 6683-6694.
24. Rathbone S, Furrer P, Lübben J, Zinn M, Cartmell S. Biocompatibility of polyhydroxyalkanoate as a potential material for ligament and tendon scaffold material. J Biomed Mater Res A. 2010; 93(4): 1391-1403.
25. Shojaee A, Parham A, Ejeian F, Nasr Esfahani MH. Equine adipose mesenchymal stem cells (eq-ASCs) appear to have higher potential for migration and musculoskeletal differentiation. Res Vet Sci. 2019; 125: 235-243.
26. Barsby T, Guest D. Transforming growth factor beta3 promotes tendon differentiation of equine embryo-derived stem cells. Tissue Eng Part A. 2013; 19(19-20): 2156-2165.
27. Winkler DG, Sutherland MS, Ojala E, Turcott E, Geoghegan JC, Shpektor D, et al. Sclerostin inhibition of Wnt-3a-induced C3H10T1/2 cell differentiation is indirect and mediated by bone morphogenetic proteins. J Biol Chem. 2005; 280(4): 2498-2502.
28. Yeung CY, Gossan N, Lu Y, Hughes A, Hensman JJ, Bayer ML, et al. Gremlin-2 is a BMP antagonist that is regulated by the circadian clock. Sci Rep. 2014; 4: 5183.
29. Masaeli E, Morshed M, Nasr-Esfahani MH, Sadri S, Hilderink J, van Apeldoorn A, et al. Fabrication, characterization and cellular compatibility of poly(hydroxy alkanoate) composite nanofibrous scaffolds for nerve tissue engineering. PLoS One. 2013; 8(2): e57157.
30. De Schauwer C, Van de Walle GR, Van Soom A, Meyer E. Mesenchymal stem cell therapy in horses: useful beyond orthopedic injuries? Vet Q. 2013; 33(4): 234-241.
31. Bavin EP, Atkinson F, Barsby T, Guest DJ. Scleraxis is essential for tendon differentiation by equine embryonic stem cells and in equine fetal tenocytes. Stem Cells Dev. 2017; 26(6): 441-450.
32. Kishimoto Y, Ohkawara B, Sakai T, Ito M, Masuda A, Ishiguro N, et al. Wnt/β-catenin signaling suppresses expressions of Scx, Mkx, and Tnmd in tendon-derived cells. PLoS One. 2017; 12(7): e0182051.
33. Soeda T, Deng JM, de Crombrugghe B, Behringer RR, Nakamura T, Akiyama H. Sox9-expressing precursors are the cellular origin of the cruciate ligament of the knee joint and the limb tendons. Genesis. 2010; 48(11): 635-644.
34. Levay AK, Peacock JD, Lu Y, Koch M, Hinton RB Jr, Kadler KE, et al. Scleraxis is required for cell lineage differentiation and extracellular matrix remodeling during murine heart valve formation in vivo. Circ Res. 2008; 103(9): 948-956.
35. Murchison ND, Price BA, Conner DA, Keene DR, Olson EN, Tabin CJ, et al. Regulation of tendon differentiation by scleraxis distinguishes force-transmitting tendons from muscle-anchoring tendons. Development. 2007; 134(14): 2697-2708.
36. Nichols AEC, Settlage RE, Werre SR, Dahlgren LA. Novel roles for scleraxis in regulating adult tenocyte function. BMC Cell Biol. 2018; 19(1): 14.
37. Shukunami C, Takimoto A, Oro M, Hiraki Y. Scleraxis positively regulates the expression of tenomodulin, a differentiation marker of tenocytes. Dev Biol. 2006; 298(1): 234-247.
38. Taylor SE, Vaughan-Thomas A, Clements DN, Pinchbeck G, Macrory LC, Smith RK, et al. Gene expression markers of tendon fibroblasts in normal and diseased tissue compared to monolayer and three dimensional culture systems. BMC Musculoskelet Disord. 2009; 10: 27.
39. Theiss F, Mirsaidi A, Mhanna R, Kümmerle J, Glanz S, Bahrenberg G, et al. Use of biomimetic microtissue spheroids and specific growth factor supplementation to improve tenocyte differentiation and adaptation to a collagen-based scaffold in vitro. Biomaterials. 2015; 69: 99-109.
40. Gomiero C, Bertolutti G, Martinello T, Van Bruaene N, Broeckx SY, Patruno M, et al. Tenogenic induction of equine mesenchymal stem cells by means of growth factors and low-level laser technology. Vet Res Commu. 2016; 40(1): 39-48.