Topography: A Biophysical Approach to Direct the Fate of Mesenchymal Stem Cells in Tissue Engineering Applications
Abstract
:1. Introduction
2. Origin and Relevance of MSCs
2.1. Isolation and Characterization
2.2. Differentiation Potential
2.3. Therapeutic Applications
3. Cell-Implant Surface Interactions
4. The Effect of Substrate Topography
4.1. Substrate Roughness
4.1.1. Ceramic-Based Scaffolds
4.1.2. Titanium (Ti)-Based Scaffolds
4.2. Substrate Patterns
4.2.1. Ceramic-Based Scaffolds
4.2.2. Ti-Based Scaffolds
4.2.3. Polymeric Substrates
4.3. Porosity
5. Conclusions and Outlook
Author Contributions
Funding
Conflicts of Interest
References
- Curtis, A.S.G.; Varde, M. Control of cell behavior: Topological factors 2. J. Natl. Cancer Inst. 1964, 33, 15–26. [Google Scholar] [CrossRef] [PubMed]
- Stevens, M.M. Exploring and engineering the cell surface interface. Science 2005, 310, 1135–1138. [Google Scholar] [CrossRef] [PubMed]
- Wu, Y.-N.; Law, J.B.K.; He, A.Y.; Low, H.Y.; Hui, J.H.; Lim, C.T.; Yang, Z.; Lee, E.H. Substrate topography determines the fate of chondrogenesis from human mesenchymal stem cells resulting in specific cartilage phenotype formation. Nanomed. Nanotechnol. Biol. Med. 2014, 10, 1507–1516. [Google Scholar] [CrossRef] [PubMed]
- Metavarayuth, K.; Sitasuwan, P.; Zhao, X.; Lin, Y.; Wang, Q. Influence of surface topographical cues on the differentiation of mesenchymal stem cells in vitro. ACS Biomater. Sci. Eng. 2016, 2, 142–151. [Google Scholar] [CrossRef]
- Brafman, D.A. Constructing stem cell microenvironments using bioengineering approaches. Physiol. Genom. 2013, 45, 1123–1135. [Google Scholar] [CrossRef] [Green Version]
- Lee, J.-H.; Park, H.-K.; Kim, K.S. Intrinsic and extrinsic mechanical properties related to the differentiation of mesenchymal stem cells. Biochem. Biophys. Res. Commun. 2016, 473, 752–757. [Google Scholar] [CrossRef]
- Moghaddam, M.M.; Bonakdar, S.; Shariatpanahi, M.R.; Shokrgozar, M.A.; Faghihi, S. The effect of physical cues on the stem cell differentiation. Curr. Stem Cell Res. Ther. 2019, 14, 268–277. [Google Scholar] [CrossRef]
- Curtis, A.; Wilkinson, C. New depths in cell behaviour: Reactions of cells to nanotopography. Biochem. Soc. Symp. 1999, 65, 15–26. [Google Scholar]
- Norman, J.J.; Desai, T.A. Methods for fabrication of nanoscale topography for tissue engineering scaffolds. Ann. Biomed. Eng. 2006, 34, 89–101. [Google Scholar] [CrossRef]
- Nie, Z.; Kumacheva, E. Patterning surfaces with functional polymers. Nat. Mater. 2008, 7, 277–290. [Google Scholar] [CrossRef]
- Dalby, M.J.; Gadegaard, N.; Herzyk, P.; Agheli, H.; Sutherland, D.S.; Wilkinson, C.D. Group analysis of regulation of fibroblast genome on low-adhesion nanostructures. Biomaterials 2007, 28, 1761–1769. [Google Scholar] [CrossRef] [PubMed]
- Nguyen, K.T.; Shukla, K.P.; Moctezuma, M.; Tang, L. Cellular and molecular responses of smooth muscle cells to surface nanotopography. J. Nanosci. Nanotechnol. 2007, 7, 2823–2832. [Google Scholar] [CrossRef] [PubMed]
- Dalby, M.J.; McCloy, D.; Robertson, M.; Agheli, H.; Sutherland, D.S.; Affrossman, S.; Oreffo, R.O. Osteoprogenitor response to semi-ordered and random nanotopographies. Biomaterials 2006, 27, 2980–2987. [Google Scholar] [CrossRef] [PubMed]
- Oh, S.; Brammer, K.S.; Li, Y.S.J.; Teng, D.; Engler, A.J.; Chien, S.; Jin, S. Stem cell fate dictated solely by altered nanotube dimension. Proc. Natl. Acad. Sci. USA 2009, 106, 2130–2135. [Google Scholar] [CrossRef] [Green Version]
- Yim, E.K.; Darling, E.M.; Kulangara, K.; Guilak, F.; Leong, K.W. Nanotopography-induced changes in focal adhesions, cytoskeletal organization, and mechanical properties of human mesenchymal stem cells. Biomaterials 2010, 31, 1299–1306. [Google Scholar] [CrossRef] [Green Version]
- Hoch, A.I.; Mittal, V.; Mitra, D.; Vollmer, N.; Zikry, C.A.; Leach, J.K. Cell-secreted matrices perpetuate the bone-forming phenotype of differentiated mesenchymal stem cells. Biomaterials 2016, 74, 178–187. [Google Scholar] [CrossRef] [Green Version]
- Leach, J.K.; Whitehead, J. Materials-directed differentiation of mesenchymal stem cells for tissue engineering and regeneration. ACS Biomater. Sci. Eng. 2017, 4, 1115–1127. [Google Scholar] [CrossRef]
- Dobbenga, S.; Fratila-Apachitei, L.E.; Zadpoor, A.A. Nanopattern-induced osteogenic differentiation of stem cells-A systematic review. Acta Biomater. 2016, 46, 3–14. [Google Scholar] [CrossRef]
- Mashinchian, O.; Turner, L.-A.; Dalby, M.J.; Laurent, S.; Shokrgozar, M.A.; Bonakdar, S.; Imani, M.; Mahmoudi, M. Regulation of stem cell fate by nanomaterial substrates. Nanomedicine 2015, 10, 829–847. [Google Scholar] [CrossRef] [Green Version]
- Engler, A.J.; Sweeney, H.L.; Discher, D.E.; Schwarzbauer, J.E. Extracellular matrix elasticity directs stem cell differentiation. J. Musculoskelet. Neuronal Interact. 2007, 7, 335. [Google Scholar]
- Engler, A.J.; Sen, S.; Sweeney, H.L.; Discher, D.E. Matrix elasticity directs stem cell lineage specification. Cell 2006, 126, 677–689. [Google Scholar] [CrossRef] [Green Version]
- Kang, E.-S.; Kim, D.-S.; Suhito, I.R.; Lee, W.; Song, I.; Kim, T.-H. Two-dimensional material-based bionano platforms to control mesenchymal stem cell differentiation. Biomater. Res. 2018, 22, 10. [Google Scholar] [CrossRef] [PubMed]
- Wang, N.; Tytell, J.D.; Ingber, D.E. Mechanotransduction at a distance: Mechanically coupling the extracellular matrix with the nucleus. Nat. Rev. Mol. Cell Biol. 2009, 10, 75–82. [Google Scholar] [CrossRef] [PubMed]
- Kim, Y.J.; Hwang, S.J.; Bae, Y.C.; Jung, J.S. miR-21 Regulates adipogenic differentiation Through the modulation of TGF-β signaling in mesenchymal stem cells derived from human adipose tissue. STEM CELLS 2009, 27, 3093–3102. [Google Scholar] [CrossRef]
- Nieden, N.I.Z.; Kempka, G.; Rancourt, D.E.; Ahr, H.-J. Induction of chondro-, osteo- and adipogenesis in embryonic stem cells by bone morphogenetic protein-2: Effect of cofactors on differentiating lineages. BMC Dev. Biol. 2005, 5, 1. [Google Scholar] [CrossRef] [Green Version]
- Derfoul, A.; Perkins, G.L.; Hall, D.J.; Tuan, R.S. Glucocorticoids promote chondrogenic differentiation of adult human mesenchymal stem cells by enhancing expression of cartilage extracellular matrix genes. STEM CELLS 2006, 24, 1487–1495. [Google Scholar] [CrossRef] [PubMed]
- Wang, J.; Chen, Z.; Sun, M.; Xu, H.; Gao, Y.; Liu, J.; Li, M. Characterization and therapeutic applications of mesenchymal stem cells for regenerative medicine. Tissue Cell 2020, 64, 101330. [Google Scholar] [CrossRef] [PubMed]
- Friedenstein, A.J.; Piatetzky-Shapiro, I.I.; Petrakova, K.V. Osteogenesis in transplants of bone marrow cells. Development 1966, 16, 381–390. [Google Scholar]
- Caplan, A.I. Mesenchymal stem cells. J. Orthop. Res. 1991, 9, 641–650. [Google Scholar] [CrossRef]
- Caplan, A.I. Mesenchymal Stem Cells: Time to Change the Name! STEM CELLS Transl. Med. 2017, 6, 1445–1451. [Google Scholar] [CrossRef] [Green Version]
- Andrzejewska, A.; Lukomska, B.; Janowski, M. Concise review: Mesenchymal stem cells: From roots to boost. STEM CELLS 2019, 37, 855–864. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bianco, P.; Robey, P.G.; Simmons, P.J. Mesenchymal stem cells: Revisiting history, concepts, and assays. Cell Stem Cell 2008, 2, 313–319. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Han, Y.; Li, X.; Zhang, Y.; Han, Y.; Chang, F.; Ding, J. Mesenchymal stem cells for regenerative medicine. Cells 2019, 8, 886. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Peng, Y.; Ke, M.; Xu, L.; Liu, L.; Chen, X.; Xia, W.; Li, X.; Chen, Z.; Ma, J.; Liao, D.; et al. Donor-derived mesenchymal stem cells combined with low-dose tacrolimus prevent acute rejection after renal transplantation. Transplantation 2013, 95, 161–168. [Google Scholar] [CrossRef]
- Marędziak, M.; Marycz, K.; Tomaszewski, K.A.; Kornicka, K.; Henry, B.M. The Influence of aging on the regenerative potential of human adipose derived mesenchymal stem cells. Stem Cells Int. 2016, 2016, 1–15. [Google Scholar] [CrossRef]
- Golpanian, S.; El-Khorazaty, J.; Mendizabal, A.; Difede, D.L.; Suncion, V.Y.; Karantalis, V.; Fishman, J.E.; Ghersin, E.; Balkan, W.; Hare, J.M. Effect of aging on human mesenchymal stem cell therapy in ischemic cardiomyopathy patients. J. Am. Coll. Cardiol. 2015, 65, 125–132. [Google Scholar] [CrossRef] [Green Version]
- Dominici, M.; Le Blanc, K.; Mueller, I.; Slaper-Cortenbach, I.; Marini, F.; Krause, D.; Deans, R.; Keating, A.; Prockop, D.; Horwitz, E. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy 2006, 8, 315–317. [Google Scholar] [CrossRef]
- Soleimani, M.; Nadri, S. A protocol for isolation and culture of mesenchymal stem cells from mouse bone marrow. Nat. Protoc. 2009, 4, 102–106. [Google Scholar] [CrossRef]
- Kim, M.J.; Shin, K.S.; Jeon, J.H.; Lee, D.R.; Shim, S.H.; Kim, J.K.; Cha, D.-H.; Yoon, T.K.; Kim, G.J. Human chorionic-plate-derived mesenchymal stem cells and Wharton’s jelly-derived mesenchymal stem cells: A comparative analysis of their potential as placenta-derived stem cells. Cell Tissue Res. 2011, 346, 53–64. [Google Scholar] [CrossRef]
- Zuk, P.A.; Zhu, M.; Ashjian, P.; De Ugarte, D.A.; Huang, J.I.; Mizuno, H.; Alfonso, Z.C.; Fraser, J.K.; Benhaim, P.; Hedrick, M.H. Human adipose tissue is a source of multipotent stem cells. Mol. Biol. Cell 2002, 13, 4279–4295. [Google Scholar] [CrossRef]
- Sibov, T.T.; Tobo, P.R.; Marti, L.C.; Pavon, L.F.; Oliveira, D.M.; Tobo, P.R.; Campos, A.H.; Paes, A.T.; Amaro, E.; Gamarra, L.F.; et al. Mesenchymal stem cells from umbilical cord blood: Parameters for isolation, characterization and adipogenic differentiation. Cytotechnology 2012, 64, 511–521. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gao, W.-X.; Sun, Y.; Shi, J.; Li, C.-L.; Fang, S.-B.; Wang, D.; Deng, X.-Q.; Wen, W.; Fu, Q.-L. Effects of mesenchymal stem cells from human induced pluripotent stem cells on differentiation, maturation, and function of dendritic cells. Stem Cell Res. Ther. 2017, 8, 48. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lin, H.; Sohn, J.; Shen, H.; Langhans, M.T.; Tuan, R.S. Bone marrow mesenchymal stem cells: Aging and tissue engineering applications to enhance bone healing. Biomaterials 2019, 203, 96–110. [Google Scholar] [CrossRef]
- Mochizuki, T.; Muneta, T.; Sakaguchi, Y.; Nimura, A.; Yokoyama, A.; Koga, H.; Sekiya, I. Higher chondrogenic potential of fibrous synovium-and adipose synovium–derived cells compared with subcutaneous fat–derived cells: Distinguishing properties of mesenchymal stem cells in humans. Arthritis Rheum. 2006, 54, 843–853. [Google Scholar] [CrossRef] [PubMed]
- Jin, H.J.; Bae, Y.K.; Kim, M.; Kwon, S.; Jeon, H.B.; Choi, S.J.; Kim, S.W.; Yang, Y.S.; Oh, W.; Chang, J.W. Comparative analysis of human mesenchymal stem cells from bone marrow, adipose tissue, and umbilical cord blood as sources of cell therapy. Int. J. Mol. Sci. 2013, 14, 17986–18001. [Google Scholar] [CrossRef] [PubMed]
- Kargozar, S.; Mozafari, M.; Hashemian, S.J.; Milan, P.B.; Hamzehlou, S.; Soleimani, M.; Joghataei, M.T.; Gholipourmalekabadi, M.; Korourian, A.; Mousavizadeh, K.; et al. Osteogenic potential of stem cells-seeded bioactive nanocomposite scaffolds: A comparative study between human mesenchymal stem cells derived from bone, umbilical cord Wharton’s jelly, and adipose tissue. J. Biomed. Mater. Res. Part B Appl. Biomater. 2016, 106, 61–72. [Google Scholar] [CrossRef]
- Burrow, K.L.; Hoyland, J.A.; Richardson, S.M. Human adipose-derived stem cells exhibit enhanced proliferative capacity and retain multipotency longer than donor-matched bone marrow mesenchymal stem cells during expansion in vitro. Stem Cells Int. 2017, 2017, 1–15. [Google Scholar] [CrossRef]
- Oreffo, R.O.; Tare, R.S.; Yang, L.-Y.; Williams, D.F.; Ou, K.-L.; Oreffo, R.O. Biofabrication of bone tissue: Approaches, challenges and translation for bone regeneration. Biomaterials 2016, 83, 363–382. [Google Scholar] [CrossRef]
- Khojasteh, A.; Fahimipour, F.; Jafarian, M.; Sharifi, D.; Jahangir, S.; Khayyatan, F.; Eslaminejad, M.B. Bone engineering in dog mandible: Coculturing mesenchymal stem cells with endothelial progenitor cells in a composite scaffold containing vascular endothelial growth factor. J. Biomed. Mater. Res. Part B Appl. Biomater. 2016, 105, 1767–1777. [Google Scholar] [CrossRef]
- Katagiri, W.; Watanabe, J.; Toyama, N.; Osugi, M.; Sakaguchi, K.; Hibi, H. Clinical study of bone regeneration by conditioned medium from mesenchymal stem cells after maxillary sinus floor elevation. Implant. Dent. 2017, 26, 607–612. [Google Scholar] [CrossRef]
- Gjerde, C.; Mustafa, K.; Hellem, S.; Rojewski, M.; Gjengedal, H.; Yassin, M.A.; Feng, X.; Skaale, S.; Berge, T.; Rosen, A.; et al. Cell therapy induced regeneration of severely atrophied mandibular bone in a clinical trial. Stem Cell Res. Ther. 2018, 9, 213. [Google Scholar] [CrossRef] [PubMed]
- Liebergall, M.; Schroeder, J.; Mosheiff, R.; Gazit, Z.; Yoram, Z.; Rasooly, L.; Daskal, A.; Khoury, A.; Weil, Y.; Beyth, S. Stem cell–based therapy for prevention of delayed fracture union: A randomized and prospective preliminary study. Mol. Ther. 2013, 21, 1631–1638. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fernandes, T.L.; Kimura, H.A.; Pinheiro, C.C.G.; Shimomura, K.; Nakamura, N.; Ferreira, J.R.; Gomoll, A.H.; Hernandez, A.J.; Bueno, D.F. Human synovial mesenchymal stem cells good manufacturing practices for articular cartilage regeneration. Tissue Eng. Part C Methods 2018, 24, 709–716. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bougioukli, S.; Sugiyama, O.; Pannell, W.; Ortega, B.; Tan, M.H.; Tang, A.H.; Yoho, R.; Oakes, D.A.; Lieberman, J.R. Gene therapy for bone repair using human cells: Superior osteogenic potential of bone morphogenetic protein 2–transduced mesenchymal stem cells derived from adipose tissue compared to bone marrow. Hum. Gene Ther. 2018, 29, 507–519. [Google Scholar] [CrossRef]
- Wakitani, S.; Goto, T.; Pineda, S.J.; Young, R.G.; Mansour, J.M.; Caplan, A.I.; Goldberg, V.M. Mesenchymal cell-based repair of large, full-thickness defects of articular cartilage. J. Bone Jt. Surg. Am. Vol. 1994, 76, 579–592. [Google Scholar] [CrossRef]
- Orozco, L.; Munar, A.; Soler, R.; Alberca, M.; Soler, F.; Huguet, M.; Sentís, J.; Sánchez, A.; García-Sancho, J. Treatment of knee osteoarthritis with autologous mesenchymal stem cells. Transplantation 2013, 95, 1535–1541. [Google Scholar] [CrossRef]
- De Windt, T.S.; Vonk, L.A.; Slaper-Cortenbach, I.C.M.; Broek, M.P.H.V.D.; Nizak, R.; Van Rijen, M.H.P.; De Weger, R.A.; Dhert, W.J.A.; Saris, D.B. Allogeneic mesenchymal stem cells stimulate cartilage regeneration and are safe for single-stage cartilage repair in humans upon mixture with recycled autologous chondrons. STEM CELLS 2016, 35, 256–264. [Google Scholar] [CrossRef]
- Orozco, L.; Soler, R.; Morera, C.; Alberca, M.; Sánchez, A.; García-Sancho, J. Intervertebral disc repair by autologous mesenchymal bone marrow cells: A pilot study. Transplant. 2011, 92, 822–828. [Google Scholar] [CrossRef] [Green Version]
- Pettine, K.A.; Suzuki, R.K.; Sand, T.T.; Murphy, M.B. Autologous bone marrow concentrate intradiscal injection for the treatment of degenerative disc disease with three-year follow-up. Int. Orthop. 2017, 41, 2097–2103. [Google Scholar] [CrossRef]
- Shapiro, S.A.; Kazmerchak, S.E.; Heckman, M.G.; Zubair, A.C.; O’Connor, M.I. A Prospective, single-blind, placebo-controlled trial of bone marrow aspirate concentrate for knee osteoarthritis. Am. J. Sports Med. 2016, 45, 82–90. [Google Scholar] [CrossRef]
- Chen, M.; Guo, W.; Gao, S.; Hao, C.; Shen, S.; Zhang, Z.; Wang, Z.; Li, X.; Jing, X.; Zhang, X.; et al. Biomechanical stimulus based strategies for meniscus tissue engineering and regeneration. Tissue Eng. Part B Rev. 2018, 24, 392–402. [Google Scholar] [CrossRef] [PubMed]
- Gülecyüz, M.F.; Macha, K.; Pietschmann, M.F.; Ficklscherer, A.; Sievers, B.; Roßbach, B.P.; Jansson, V.; Müller, P.E. Allogenic myocytes and mesenchymal stem cells partially improve fatty rotator cuff degeneration in a rat model. Stem Cell Rev. Rep. 2018, 14, 847–859. [Google Scholar] [CrossRef] [PubMed]
- Bae, K.S.; Park, J.B.; Kim, H.S.; Kim, D.S.; Park, D.J.; Kang, S.J. Neuron-like differentiation of bone marrow-derived mesenchymal stem cells. Yonsei Med. J. 2011, 52, 401–412. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Oh, S.K.; Choi, K.H.; Yoo, J.Y.; Kim, D.Y.; Kim, S.J.; Jeon, S.R. A Phase III clinical trial showing limited efficacy of autologous mesenchymal stem cell therapy for spinal cord injury. Neurosurgery 2015, 78, 436–447. [Google Scholar] [CrossRef] [Green Version]
- Wang, S.; Cheng, H.; Dai, G.; Wang, X.; Hua, R.; Liu, X.; Wang, P.; Chen, G.; Yue, W.; An, Y. Umbilical cord mesenchymal stem cell transplantation significantly improves neurological function in patients with sequelae of traumatic brain injury. Brain Res. 2013, 1532, 76–84. [Google Scholar] [CrossRef]
- Harris, V.K.; Stark, J.; Vyshkina, T.; Blackshear, L.; Joo, G.; Stefanova, V.; Sara, G.; Sadiq, S.A. Phase I trial of intrathecal mesenchymal stem cell-derived neural progenitors in progressive multiple sclerosis. EBioMedicine 2018, 29, 23–30. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shichinohe, H.; Kawabori, M.; Iijima, H.; Teramoto, T.; Abumiya, T.; Nakayama, N.; Kazumata, K.; Terasaka, S.; Arato, T.; Houkin, K. Research on advanced intervention using novel bone marrOW stem cell (RAINBOW): A study protocol for a phase I, open-label, uncontrolled, dose-response trial of autologous bone marrow stromal cell transplantation in patients with acute ischemic stroke. BMC Neurol. 2017, 17, 1–8. [Google Scholar] [CrossRef]
- Venkatesh, K.; Sen, D. mesenchymal stem cells as a source of dopaminergic neurons: A potential cell based therapy for Parkinson’s disease. Curr. Stem Cell Res. Ther. 2017, 12, 326–347. [Google Scholar] [CrossRef]
- Tse, H.-F.; Kwong, Y.-L.; Chan, J.K.F.; Lo, G.; Ho, C.-L.; Lau, C.-P. Angiogenesis in ischaemic myocardium by intramyocardial autologous bone marrow mononuclear cell implantation. Lancet 2003, 361, 47–49. [Google Scholar] [CrossRef]
- Aggarwal, S.; Pittenger, M.F. Human mesenchymal stem cells modulate allogeneic immune cell responses. Blood 2005, 105, 1815–1822. [Google Scholar] [CrossRef] [Green Version]
- Petersen, B.E. Bone marrow as a potential source of hepatic oval cells. Science 1999, 284, 1168–1170. [Google Scholar] [CrossRef] [PubMed]
- Von Der Mark, K.; Park, J. Engineering biocompatible implant surfaces. Prog. Mater. Sci. 2013, 58, 327–381. [Google Scholar] [CrossRef]
- Lehnert, D.; Wehrle-Haller, B.; David, C.; Weiland, U.; Ballestrem, C.; Imhof, B.A.; Bastmeyer, M. Cell behaviour on micropatterned substrata: Limits of extracellular matrix geometry for spreading and adhesion. J. Cell Sci. 2004, 117, 41–52. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, X.; Fan, H.; Deng, X.; Wu, L.; Yi, T.; Gu, L.; Zhou, C.; Fan, Y.; Zhang, X. Scaffold structural microenvironmental cues to guide tissue regeneration in bone tissue applications. Nanomaterials 2018, 8, 960. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hynes, R.O. Integrins. Cell 2002, 110, 673–687. [Google Scholar] [CrossRef] [Green Version]
- Arima, Y.; Iwata, H. Effect of wettability and surface functional groups on protein adsorption and cell adhesion using well-defined mixed self-assembled monolayers. Biomaterials 2007, 28, 3074–3082. [Google Scholar] [CrossRef]
- Zhu, X.; Fan, H.; Xiao, Y.; Li, D.; Zhang, H.; Luxbacher, T.; Zhang, X. Effect of surface structure on protein adsorption to biphasic calcium-phosphate ceramics in vitro and in vivo. Acta Biomater. 2009, 5, 1311–1318. [Google Scholar] [CrossRef]
- Higgins, A.M.; Brown, J.L. Osteoinductive biomaterial geometries for bone regenerative engineering. Curr. Pharm. Des. 2013, 19, 3446–3455. [Google Scholar] [CrossRef]
- Vogel, V.; Sheetz, M. Local force and geometry sensing regulate cell functions. Nat. Rev. Mol. Cell Biol. 2006, 7, 265–275. [Google Scholar] [CrossRef] [PubMed]
- Cukierman, E.; Pankov, R.; Stevens, D.R.; Yamada, K.M. Taking cell-matrix adhesions to the third dimension. Science 2001, 294, 1708–1712. [Google Scholar] [CrossRef] [PubMed]
- McBeath, R.; Pirone, D.M.; Nelson, C.M.; Bhadriraju, K.; Chen, C.S. Cell Shape, cytoskeletal tension, and RhoA regulate stem cell lineage commitment. Dev. Cell 2004, 6, 483–495. [Google Scholar] [CrossRef] [Green Version]
- Lavenus, S.; Ricquier, J.-C.; Louarn, G.; Layrolle, P. Cell interaction with nanopatterned surface of implants. Nanomedicine 2010, 5, 937–947. [Google Scholar] [CrossRef] [PubMed]
- Zhu, X.; Chen, J.; Scheideler, L.; Altebaeumer, T.; Geis-Gerstorfer, J.; Kern, D. Cellular reactions of osteoblasts to micron-and submicron-scale porous structures of titanium surfaces. Cells Tissues Organs 2004, 178, 13–22. [Google Scholar] [CrossRef] [PubMed]
- Limat, A.; Hunziker, T.; Breitkreutz, D.; Fusenig, N.E.; Braathen, L.R. Organotypic cocultures as models to study cell-cell and cell-matrix interactions of human hair follicle cells. Ski. Pharmacol. Physiol. 1994, 7, 47–54. [Google Scholar] [CrossRef]
- Giancotti, F.G. Integrin signaling. Science 1999, 285, 1028–1033. [Google Scholar] [CrossRef] [PubMed]
- McNamara, L.E.; McMurray, R.J.; Biggs, M.J.P.; Kantawong, F.; Oreffo, R.O.; Dalby, M.J. Nanotopographical control of stem cell differentiation. J. Tissue Eng. 2010, 2010, 120623. [Google Scholar] [CrossRef]
- Harrison, R.G. On the stereotropism of embryonic cells. Science 1911, 34, 279–281. [Google Scholar] [CrossRef] [Green Version]
- Lozano-Calderón, S.A.; Swaim, S.O.; Federico, A.; Anderson, M.E.; Gebhardt, M.C. Predictors of soft-tissue complications and deep infection in allograft reconstruction of the proximal tibia. J. Surg. Oncol. 2016, 113, 811–817. [Google Scholar] [CrossRef]
- Jiang, Y.; Jahagirdar, B.N.; Reinhardt, R.L.; Schwartz, R.E.; Keene, C.D.; Ortiz-Gonzalez, X.R.; Reyes, M.; Lenvik, T.; Lund, T.; Blackstad, M.; et al. Pluripotency of mesenchymal stem cells derived from adult marrow. Nat. Cell Biol. 2002, 418, 41–49. [Google Scholar] [CrossRef] [Green Version]
- Yang, W.; Han, W.; He, W.; Li, J.; Wang, J.; Feng, H.; Qian, Y. Surface topography of hydroxyapatite promotes osteogenic differentiation of human bone marrow mesenchymal stem cells. Mater. Sci. Eng. C 2016, 60, 45–53. [Google Scholar] [CrossRef]
- Matsuzaka, K.; Yoshinari, M.; Shimono, M.; Inoue, T. Effects of multigrooved surfaces on osteoblast-like cellsin vitro: Scanning electron microscopic observation and mRNA expression of osteopontin and osteocalcin. J. Biomed. Mater. Res. 2003, 68, 227–234. [Google Scholar] [CrossRef]
- Faia-Torres, A.B.; Guimond-Lischer, S.; Rottmar, M.; Charnley, M.; Goren, T.; Maniura-Weber, K.; Spencer, N.D.; Reis, R.L.; Textor, M.; Neves, N.M. Differential regulation of osteogenic differentiation of stem cells on surface roughness gradients. Biomaterials 2014, 35, 9023–9032. [Google Scholar] [CrossRef] [PubMed]
- Hefti, T.; Frischherz, M.; Spencer, N.D.; Hall, H.; Schlottig, F. A comparison of osteoclast resorption pits on bone with titanium and zirconia surfaces. Biomaterials 2010, 31, 7321–7331. [Google Scholar] [CrossRef]
- Faia-Torres, A.B.; Charnley, M.; Goren, T.; Guimond-Lischer, S.; Rottmar, M.; Maniura-Weber, K.; Spencer, N.D.; Reis, R.L.; Textor, M.; Neves, N.M. Osteogenic differentiation of human mesenchymal stem cells in the absence of osteogenic supplements: A surface-roughness gradient study. Acta Biomater. 2015, 28, 64–75. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hong, J.-H.; Wilson, W.H.; Schroeder, D.C.; Allen, M.J.; Holden, M.T.G.; Parkhill, J.; Barrell, B.G.; Churcher, C.; Hamlin, N.; Mungall, K.; et al. TAZ, a Transcriptional modulator of mesenchymal stem cell differentiation. Science 2005, 309, 1074–1078. [Google Scholar] [CrossRef] [Green Version]
- Dupont, S.; Morsut, L.; Aragona, M.; Enzo, E.; Giulitti, S.; Cordenonsi, M.; Zanconato, F.; Le Digabel, J.; Forcato, M.; Bicciato, S.; et al. Role of YAP/TAZ in mechanotransduction. Nat. Cell Biol. 2011, 474, 179–183. [Google Scholar] [CrossRef] [PubMed]
- Schantz, J.-T.; Lim, T.C.; Ning, C.; Teoh, S.H.; Tan, K.C.; Wang, S.C.; Hutmacher, D.W. Cranioplasty after trephination using a novel biodegradable burr hole cover: Technical case report. Oper. Neurosurg. 2006, 58. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hou, Y.; Xie, W.; Yu, L.; Camacho, L.C.; Nie, C.; Zhang, M.; Haag, R.; Wei, Q. Surface roughness gradients reveal topography-specific mechanosensitive responses in human mesenchymal stem cells. Small 2020, 16, e1905422. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Greer, A.I.; Goriainov, V.; Kanczler, J.; Black, C.R.; Turner, L.-A.; Meek, R.M.; Burgess, K.; MacLaren, I.; Dalby, M.J.; Oreffo, R.O.; et al. Nanopatterned titanium implants accelerate bone formation in vivo. ACS Appl. Mater. Interfaces 2020, 12, 33541–33549. [Google Scholar] [CrossRef]
- Sjöström, T.; McNamara, L.E.; Dalby, M.J.; Meek, R.M.D.; Su, B. 2D and 3D Nanopatterning of titanium for enhancing osteoinduction of stem cells at implant surfaces. Adv. Heal. Mater. 2013, 2, 1285–1293. [Google Scholar] [CrossRef] [PubMed]
- Chen, W.; Xu, K.; Tao, B.; Dai, L.; Yu, Y.; Mu, C.; Shen, X.; Hu, Y.; He, Y.; Cai, K. Multilayered coating of titanium implants promotes coupled osteogenesis and angiogenesis in vitro and in vivo. Acta Biomater. 2018, 74, 489–504. [Google Scholar] [CrossRef] [PubMed]
- Nelson, K.L.; Cox, M.D.; Richter, G.T.; Dornhoffer, J.L. A comparative review of osseointegration failure between osseointegrated bone conduction device models in pediatric patients. Otol. Neurotol. 2016, 37, 276–280. [Google Scholar] [CrossRef] [PubMed]
- Sjöström, T.; Dalby, M.J.; Hart, A.; Tare, R.; Oreffo, R.O.; Su, B. Fabrication of pillar-like titania nanostructures on titanium and their interactions with human skeletal stem cells. Acta Biomater. 2009, 5, 1433–1441. [Google Scholar] [CrossRef] [PubMed]
- Khang, D.; Choi, J.; Im, Y.-M.; Kim, Y.-J.; Jang, J.-H.; Kang, S.S.; Nam, T.-H.; Song, J.; Park, J.-W. Role of subnano-, nano- and submicron-surface features on osteoblast differentiation of bone marrow mesenchymal stem cells. Biomaterials 2012, 33, 5997–6007. [Google Scholar] [CrossRef] [PubMed]
- Cai, K.; Bossert, J.; Jandt, K.D. Does the nanometre scale topography of titanium influence protein adsorption and cell proliferation? Colloids Surf. B Biointerfaces 2006, 49, 136–144. [Google Scholar] [CrossRef]
- Xu, K.; Chen, W.; Hu, Y.; Shen, X.; Xu, G.; Ran, Q.; Yu, Y.; Mu, C.; Cai, K. Influence of strontium ions incorporated into nanosheet-pore topographical titanium substrates on osteogenic differentiation of mesenchymal stem cells in vitro and on osseointegration in vivo. J. Mater. Chem. B 2016, 4, 4549–4564. [Google Scholar] [CrossRef]
- Xu, K.; Shen, X.; Chen, W.; Mu, C.; Jiang, C.; Zhao, Y.; Cai, K. Nanosheet-pore topographical titanium substrates: A biophysical regulator of the fate of mesenchymal stem cells. J. Mater. Chem. B 2016, 4, 1797–1810. [Google Scholar] [CrossRef]
- Zhao, L.; Wang, H.; Huo, K.; Zhang, X.; Wang, W.; Zhang, Y.; Wu, Z.; Chu, P.K. The osteogenic activity of strontium loaded titania nanotube arrays on titanium substrates. Biomaterials 2013, 34, 19–29. [Google Scholar] [CrossRef]
- Huang, Y.; Zha, G.; Luo, Q.; Zhang, J.; Zhang, F.; Li, X.; Zhao, S.; Zhu, W.; Li, X. The construction of hierarchical structure on Ti substrate with superior osteogenic activity and intrinsic antibacterial capability. Sci. Rep. 2014, 4, 6172. [Google Scholar] [CrossRef] [Green Version]
- Heitz-Mayfield, L.J. Peri-implant diseases: Diagnosis and risk indicators. J. Clin. Periodontol. 2008, 35, 292–304. [Google Scholar] [CrossRef]
- Lotz, E.M.; Berger, M.B.; Schwartz, Z.; Boyan, B.D. Regulation of osteoclasts by osteoblast lineage cells depends on titanium implant surface properties. Acta Biomater. 2018, 68, 296–307. [Google Scholar] [CrossRef] [PubMed]
- Zhao, C.; Wang, X.; Gao, L.; Jing, L.; Zhou, Q.; Chang, J. The role of the micro-pattern and nano-topography of hydroxyapatite bioceramics on stimulating osteogenic differentiation of mesenchymal stem cells. Acta Biomater. 2018, 73, 509–521. [Google Scholar] [CrossRef] [PubMed]
- Hu, C.; Ashok, D.; Nisbet, D.R.; Gautam, V. Bioinspired surface modification of orthopedic implants for bone tissue engineering. Biomaterials 2019, 219, 119366. [Google Scholar] [CrossRef]
- Xia, L.; Lin, K.; Jiang, X.; Xu, Y.; Zhang, M.; Chang, J.; Zhang, Z. Enhanced osteogenesis through nano-structured surface design of macroporous hydroxyapatite bioceramic scaffolds via activation of ERK and p38 MAPK signaling pathways. J. Mater. Chem. B 2013, 1, 5403–5416. [Google Scholar] [CrossRef]
- Yuan, H.; Fernandes, H.; Habibovic, P.; De Boer, J.; Barradas, A.M.C.; De Ruiter, A.; Walsh, W.R.; Van Blitterswijk, C.A.; De Bruijn, J.D. Osteoinductive ceramics as a synthetic alternative to autologous bone grafting. Proc. Natl. Acad. Sci. USA 2010, 107, 13614–13619. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, C.; Fan, Z.; Han, Y. Formation and osteoblast behavior of HA nano-rod/fiber patterned coatings on tantalum in porous and compact forms. J. Mater. Chem. B 2015, 3, 5442–5454. [Google Scholar] [CrossRef] [PubMed]
- Zhou, J.; Li, B.; Lu, S.; Zhang, L.; Han, Y. Regulation of osteoblast proliferation and differentiation by interrod spacing of Sr-HA nanorods on microporous titania coatings. ACS Appl. Mater. Interf. 2013, 5, 5358–5365. [Google Scholar] [CrossRef] [PubMed]
- Zhou, J.; Li, B.; Han, Y.; Zhao, L. The osteogenic capacity of biomimetic hierarchical micropore/nanorod-patterned Sr-HA coatings with different interrod spacings. Nanomed. Nanotechnol. Biol. Med. 2016, 12, 1161–1173. [Google Scholar] [CrossRef] [PubMed]
- Zhou, J.; Zhao, L.; Li, B.; Han, Y. Nanorod diameter modulated osteogenic activity of hierarchical micropore/nanorod-patterned coatings via a Wnt/β-catenin pathway. Nanomed. Nanotechnol. Biol. Med. 2018, 14, 1719–1731. [Google Scholar] [CrossRef]
- Loye, A.M.; Kinser, E.R.; Bensouda, S.; Shayan, M.; Davis, R.; Wang, R.; Chen, Z.; Schwarz, U.D.; Schroers, J.; Kyriakides, T.R. Regulation of mesenchymal stem cell differentiation by nanopatterning of bulk metallic glass. Sci. Rep. 2018, 8, 8758. [Google Scholar] [CrossRef]
- Kumar, G.; Tang, H.X.; Schroers, J. Nanomoulding with amorphous metals. Nat. Cell Biol. 2009, 457, 868–872. [Google Scholar] [CrossRef] [PubMed]
- Kumar, G.; Staffier, P.A.; Blawzdziewicz, J.; Schwarz, U.D.; Schroers, J. Atomically smooth surfaces through thermoplastic forming of metallic glass. Appl. Phys. Lett. 2010, 97, 101907. [Google Scholar] [CrossRef] [Green Version]
- Kumar, G.; Blawzdziewicz, J.; Schroers, J. Controllable nanoimprinting of metallic glasses: Effect of pressure and interfacial properties. Nanotechnology 2013, 24, 105301. [Google Scholar] [CrossRef] [PubMed]
- Wang, W.; Dong, C.; Shek, C. Bulk metallic glasses. Mater. Sci. Eng. R Rep. 2004, 44, 45–89. [Google Scholar] [CrossRef]
- Lee, D.-K.; Yi, T.; Park, K.-E.; Lee, H.-J.; Cho, Y.-K.; Lee, S.J.; Lee, J.; Park, J.H.; Lee, M.-Y.; Song, S.U.; et al. Non-invasive characterization of the adipogenic differentiation of human bone marrow-derived mesenchymal stromal cells by HS-SPME/GC-MS. Sci. Rep. 2014, 4, 6550. [Google Scholar] [CrossRef]
- Lin, K.; Xia, L.; Zhai, D.; Zhang, N.; Liu, J.; Fang, B.; Chang, J.; Lin, K. Designing ordered micropatterned hydroxyapatite bioceramics to promote the growth and osteogenic differentiation of bone marrow stromal cells. J. Mater. Chem. B 2015, 3, 968–976. [Google Scholar] [CrossRef]
- Yang, C.; Zhao, C.; Wang, X.; Shi, M.; Zhu, Y.; Jing, L.; Wu, C.; Chang, J. Stimulation of osteogenesis and angiogenesis by micro/nano hierarchical hydroxyapatite via macrophage immunomodulation. Nanoscale 2019, 11, 17699–17708. [Google Scholar] [CrossRef]
- Xia, L.; Lin, K.; Jiang, X.; Fang, B.; Xu, Y.; Liu, J.; Zeng, D.; Zhang, M.; Zhang, X.; Chang, J.; et al. Effect of nano-structured bioceramic surface on osteogenic differentiation of adipose derived stem cells. Biomaterials 2014, 35, 8514–8527. [Google Scholar] [CrossRef]
- Lin, K.; Xia, L.; Gan, J.; Zhang, Z.; Chen, H.; Jiang, X.; Chang, J. Tailoring the nanostructured surfaces of hydroxyapatite bioceramics to promote protein adsorption, osteoblast growth, and osteogenic differentiation. ACS Appl. Mater. Interf. 2013, 5, 8008–8017. [Google Scholar] [CrossRef]
- Spiller, K.L.; Koh, T.J. Macrophage-based therapeutic strategies in regenerative medicine. Adv. Drug Deliv. Rev. 2017, 122, 74–83. [Google Scholar] [CrossRef]
- Sridharan, R.; Cameron, A.R.; Kelly, D.J.; Kearney, C.J.; O’Brien, F.J. Biomaterial based modulation of macrophage polarization: A review and suggested design principles. Mater. Today 2015, 18, 313–325. [Google Scholar] [CrossRef]
- Lee, C.Z.W.; Kozaki, T.; Ginhoux, F. Studying tissue macrophages in vitro: Are iPSC-derived cells the answer? Nat. Rev. Immunol. 2018, 18, 716–725. [Google Scholar] [CrossRef] [PubMed]
- Alaarg, A.; Pérez-Medina, C.; Metselaar, J.M.; Nahrendorf, M.; Fayad, Z.A.; Storm, G.; Mulder, W.J.M. Applying nanomedicine in maladaptive inflammation and angiogenesis. Adv. Drug Deliv. Rev. 2017, 119, 143–158. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Q.; Hwang, J.W.; Oh, J.-H.; Park, C.H.; Chung, S.H.; Lee, Y.-S.; Baek, J.-H.; Ryoo, H.-M.; Woo, K.M. Effects of the fibrous topography-mediated macrophage phenotype transition on the recruitment of mesenchymal stem cells: An in vivo study. Biomaterials 2017, 149, 77–87. [Google Scholar] [CrossRef]
- Mukherjee, S.; Darzi, S.; Rosamilia, A.; Kadam, V.; Truong, Y.B.; Werkmeister, J.A.; Gargett, C.E. Blended nanostructured degradable mesh with endometrial mesenchymal stem cells promotes tissue integration and anti-inflammatory response in vivo for pelvic floor application. Biomacromolecules 2018, 20, 454–468. [Google Scholar] [CrossRef]
- Li, K.; Hu, D.; Xie, Y.; Huang, L.; Zheng, X. Sr-doped nanowire modification of Ca–Si-based coatings for improved osteogenic activities and reduced inflammatory reactions. Nanotechnology 2018, 29, 084001. [Google Scholar] [CrossRef]
- Zhang, Q.; Ma, L.; Zheng, S.; Wang, Y.; Feng, M.; Shuai, Y.; Duan, B.; Fan, X.; Yang, M.; Mao, C. Air-plasma treatment promotes bone-like nano-hydroxylapatite formation on protein films for enhanced in vivo osteogenesis. Biomater. Sci. 2019, 7, 2326–2334. [Google Scholar] [CrossRef]
- Lin, K.-F.; He, S.; Song, Y.; Wang, C.-M.; Gao, Y.; Li, J.-Q.; Tang, P.; Wang, Z.; Bi, L.; Pei, G.-X. Low-temperature additive manufacturing of biomimic three-dimensional hydroxyapatite/collagen scaffolds for bone regeneration. ACS Appl. Mater. Interf. 2016, 8, 6905–6916. [Google Scholar] [CrossRef]
- Wagener, V.; Boccaccini, A.R.; Virtanen, S. Protein-adsorption and Ca-phosphate formation on chitosan-bioactive glass composite coatings. Appl. Surf. Sci. 2017, 416, 454–460. [Google Scholar] [CrossRef]
- Cardoso, G.B.C.; Maniglio, D.; Volpato, F.Z.F.Z.; Tondon, A.; Migliaresi, C.; Kaunas, R.; Zavaglia, C.A.C.C.A.C. Oleic acid surfactant in polycaprolactone/hydroxyapatite-composites for bone tissue engineering. J. Biomed. Mater. Res. Part B Appl. Biomater. 2015, 104, 1076–1082. [Google Scholar] [CrossRef]
- Gogoi, D.; Choudhury, A.J.; Chutia, J.; Pal, A.R.; Khan, M.; Choudhury, M.; Pathak, P.; Das, G.; Patil, D.S. Development of advanced antimicrobial and sterilized plasma polypropylene grafted muga (antheraea assama) silk as suture biomaterial. Biopolymers 2014, 101, 355–365. [Google Scholar] [CrossRef] [PubMed]
- Jiang, J.; Hao, W.; Li, Y.; Yao, J.; Zheng, G.; Li, H.; Yang, J.; Chen, S. Erratum to: Hydroxyapatite/regenerated silk fibroin scaffold-enhanced osteoinductivity and osteoconductivity of bone marrow-derived mesenchymal stromal cells. Biotechnol. Lett. 2013, 35, 1349–1350. [Google Scholar] [CrossRef] [Green Version]
- Liu, H.; Xu, G.W.; Wang, Y.F.; Zhao, H.S.; Xiong, S.; Wu, Y.; Heng, B.C.; An, C.R.; Zhu, G.H.; Xie, D.H. Composite scaffolds of nano-hydroxyapatite and silk fibroin enhance mesenchymal stem cell-based bone regeneration via the interleukin 1 alpha autocrine/paracrine signaling loop. Biomaterials 2015, 49, 103–112. [Google Scholar] [CrossRef] [PubMed]
- Wang, N.; Li, H.; Lü, W.; Li, J.; Wang, J.; Zhang, Z.; Liu, Y. Effects of TiO2 nanotubes with different diameters on gene expression and osseointegration of implants in minipigs. Biomaterials 2011, 32, 6900–6911. [Google Scholar] [CrossRef]
- Qiu, J.; Li, J.; Wang, S.; Ma, B.; Zhang, S.; Guo, W.; Zhang, X.; Tang, W.; Sang, Y.; Liu, H. TiO2 nanorod array constructed nanotopography for regulation of mesenchymal stem cells fate and the realization of location-committed stem cell differentiation. Small 2016, 12, 1770–1778. [Google Scholar] [CrossRef] [PubMed]
- Lv, L.; Liu, Y.; Zhang, P.; Zhang, X.; Liu, J.; Chen, T.; Su, P.; Li, H.; Zhou, Y. The nanoscale geometry of TiO2 nanotubes influences the osteogenic differentiation of human adipose-derived stem cells by modulating H3K4 trimethylation. Biomaterials 2015, 39, 193–205. [Google Scholar] [CrossRef] [PubMed]
- Lavenus, S.; Trichet, V.; Le Chevalier, S.; Hoornaert, A.; Louarn, G.; Layrolle, P. Cell differentiation and osseointegration influenced by nanoscale anodized titanium surfaces. Nanomedicine 2012, 7, 967–980. [Google Scholar] [CrossRef]
- Oh, S.; Daraio, C.; Chen, L.-H.; Pisanic, T.R.; Fiñones, R.R.; Jin, S. Significantly accelerated osteoblast cell growth on aligned TiO2 nanotubes. J. Biomed. Mater. Res. Part A 2006, 78, 97–103. [Google Scholar] [CrossRef]
- Hou, W.; Fu, H.; Liu, X.; Duan, K.; Lu, X.; Lu, M.; Sun, T.; Guo, T.; Weng, J. Cation Channel Transient Receptor Potential Vanilloid 4 Mediates Topography-Induced Osteoblastic Differentiation of Bone Marrow Stem Cells. ACS Biomater. Sci. Eng. 2019, 5, 6520–6529. [Google Scholar] [CrossRef]
- Lee, H.-P.; Stowers, R.; Chaudhuri, O. Volume expansion and TRPV4 activation regulate stem cell fate in three-dimensional microenvironments. Nat. Commun. 2019, 10, 529. [Google Scholar] [CrossRef]
- Jiang, X.; Zhang, W.; Li, Z.; Xu, L.; Li, J.; Jin, Y.; Wang, G.; Liu, X.; Huang, Q. Effects of a hybrid micro/nanorod topography-modified titanium implant on adhesion and osteogenic differentiation in rat bone marrow mesenchymal stem cells. Int. J. Nanomed. 2013, 8, 257–265. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dalby, M.J.; Gadegaard, N.; Tare, R.; Andar, A.; Riehle, M.O.; Herzyk, P.; Wilkinson, C.D.W.; Oreffo, R.O.C. The control of human mesenchymal cell differentiation using nanoscale symmetry and disorder. Nat. Mater. 2007, 6, 997–1003. [Google Scholar] [CrossRef] [PubMed]
- McMurray, R.J.; Gadegaard, N.; Tsimbouri, P.M.; Burgess, K.V.; McNamara, L.E.; Tare, R.; Murawski, K.; Kingham, E.; Oreffo, R.O.C.; Dalby, M.J. Nanoscale surfaces for the long-term maintenance of mesenchymal stem cell phenotype and multipotency. Nat. Mater. 2011, 10, 637–644. [Google Scholar] [CrossRef] [PubMed]
- Abagnale, G.; Steger, M.; Nguyen, V.H.; Hersch, N.; Sechi, A.; Joussen, S.; Denecke, B.; Merkel, R.; Hoffmann, B.; Dreser, A.; et al. Surface topography enhances differentiation of mesenchymal stem cells towards osteogenic and adipogenic lineages. Biomaterials 2015, 61, 316–326. [Google Scholar] [CrossRef] [PubMed]
- Rubehn, B.; Stieglitz, T. In vitro evaluation of the long-term stability of polyimide as a material for neural implants. Biomaterials 2010, 31, 3449–3458. [Google Scholar] [CrossRef]
- Sun, Y.; Lacour, S.P.; Brooks, R.A.; Rushton, N.; Fawcett, J.W.; Cameron, R.E. Assessment of the biocompatibility of photosensitive polyimide for implantable medical device use. J. Biomed. Mater. Res. Part A 2009, 90, 648–655. [Google Scholar] [CrossRef]
- Prichard, H.L.; Reichert, W.M.; Klitzman, B. Adult adipose-derived stem cell attachment to biomaterials. Biomaterials 2007, 28, 936–946. [Google Scholar] [CrossRef] [Green Version]
- Kolambkar, Y.M.; Bajin, M.; Wojtowicz, A.; Hutmacher, D.W.; Garcia, A.J.; Guldberg, R.E. Nanofiber orientation and surface functionalization modulate human mesenchymal stem cell behavior in vitro. Tissue Eng. Part A 2013, 20, 398–409. [Google Scholar] [CrossRef] [Green Version]
- Polini, A.; Pisignano, D.; Parodi, M.; Quarto, R.; Scaglione, S. Osteoinduction of human mesenchymal stem cells by bioactive composite scaffolds without supplemental osteogenic growth factors. PLoS ONE 2011, 6, e26211. [Google Scholar] [CrossRef]
- Lü, L.-X.; Wang, Y.-Y.; Mao, X.; Xiao, Z.; Huang, N.-P. The effects of PHBV electrospun fibers with different diameters and orientations on growth behavior of bone-marrow-derived mesenchymal stem cells. Biomed. Mater. 2012, 7, 015002. [Google Scholar] [CrossRef]
- Lü, L.-X.; Zhang, X.-F.; Wang, Y.-Y.; Ortiz, L.; Mao, X.; Jiang, Z.-L.; Xiao, Z.-D.; Huang, N.-P. Effects of hydroxyapatite-containing composite nanofibers on osteogenesis of mesenchymal stem cells in vitro and bone regeneration in vivo. ACS Appl. Mater. Interf. 2013, 5, 319–330. [Google Scholar] [CrossRef]
- Zhang, N.; Xiao, Q.-R.; Man, X.-Y.; Liu, H.-X.; Lü, L.-X.; Huang, N.-P. Spontaneous osteogenic differentiation of mesenchymal stem cells on electrospun nanofibrous scaffolds. RSC Adv. 2016, 6, 22144–22152. [Google Scholar] [CrossRef]
- Liu, F.; Kohlmeier, S.; Wang, C.-Y. Wnt signaling and skeletal development. Cell. Signal. 2008, 20, 999–1009. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Guan, J.; Zhang, J.; Guo, S.; Zhu, H.; Zhu, Z.; Li, H.; Wang, Y.; Zhang, C.; Chang, J. Human urine-derived stem cells can be induced into osteogenic lineage by silicate bioceramics via activation of the Wnt/β-catenin signaling pathway. Biomaterials 2015, 55, 1–11. [Google Scholar] [CrossRef]
- Nishimura, R.; Hata, K.; Harris, S.; Ikeda, F.; Yoneda, T. Core-binding factor α1 (Cbfa1) induces osteoblastic differentiation of C2C12 cells without interactions with Smad1 and Smad5. Bone 2002, 31, 303–312. [Google Scholar] [CrossRef]
- Wang, W.; Liu, Q.; Zhang, Y.; Zhao, L. Involvement of ILK/ERK1/2 and ILK/p38 pathways in mediating the enhanced osteoblast differentiation by micro/nanotopography. Acta Biomater. 2014, 10, 3705–3715. [Google Scholar] [CrossRef] [PubMed]
- Izadpanahi, M.; Seyedjafari, E.; Arefian, E.; Hamta, A.; Hosseinzadeh, S.; Kehtari, M.; Soleimani, M.; Kehtari, M. Nanotopographical cues of electrospun PLLA efficiently modulate non-coding RNA network to osteogenic differentiation of mesenchymal stem cells during BMP signaling pathway. Mater. Sci. Eng. C 2018, 93, 686–703. [Google Scholar] [CrossRef]
- Kaplan, L.; Fu, F. Clinical Applications of Orthopedic Tissue Engineering. Orthop. Tissue Eng. 2004, 261. [Google Scholar] [CrossRef]
- Teh, T.K.; Toh, S.-L.; Goh, J.C. Aligned fibrous scaffolds for enhanced mechanoresponse and tenogenesis of mesenchymal stem cells. Tissue Eng. Part A 2013, 19, 1360–1372. [Google Scholar] [CrossRef]
- Woo, S.L.-Y.; Debski, R.E.; Zeminski, J.; Abramowitch, S.D.; Saw, S.; Fenwick, J.A. Injury and repair of ligaments and tendons. Annu. Rev. Biomed. Eng. 2000, 2, 83–118. [Google Scholar] [CrossRef]
- Woo, S.L.-Y.; Vogrin, T.M.; Abramowitch, S.D. Healing and repair of ligament injuries in the knee. J. Am. Acad. Orthop. Surg. 2000, 8, 364–372. [Google Scholar] [CrossRef]
- Hogan, M.V.; Bagayoko, N.; James, R.; Starnes, T.; Katz, A.; Chhabra, B.A. Tissue engineering solutions for tendon repair. J. Am. Acad. Orthop. Surg. 2011, 19, 134–142. [Google Scholar] [CrossRef] [PubMed]
- Czaplewski, S.K.; Tsai, T.-L.; Duenwald-Kuehl, S.E.; Vanderby, R.; Li, W.-J. Tenogenic differentiation of human induced pluripotent stem cell-derived mesenchymal stem cells dictated by properties of braided submicron fibrous scaffolds. Biomaterials 2014, 35, 6907–6917. [Google Scholar] [CrossRef]
- Ma, Z.; Kotaki, M.; Inai, R.; Ramakrishna, S. Potential of nanofiber matrix as tissue-engineering scaffolds. Tissue Eng. 2005, 11, 101–109. [Google Scholar] [CrossRef] [PubMed]
- Yin, Z.; Chen, X.; Chen, J.L.; Shen, W.-L.; Nguyen, T.M.H.; Gao, L.; Ouyang, H.W. The regulation of tendon stem cell differentiation by the alignment of nanofibers. Biomaterials 2010, 31, 2163–2175. [Google Scholar] [CrossRef] [PubMed]
- Kishore, V.; Bullock, W.; Sun, X.; Van Dyke, W.S.; Akkus, O. Tenogenic differentiation of human MSCs induced by the topography of electrochemically aligned collagen threads. Biomaterials 2012, 33, 2137–2144. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tong, W.Y.; Shen, W.; Yeung, C.W.; Zhao, Y.; Cheng, S.H.; Chu, P.K.; Chan, D.; Chan, G.C.; Cheung, K.M.; Yeung, K.W.K.; et al. Functional replication of the tendon tissue microenvironment by a bioimprinted substrate and the support of tenocytic differentiation of mesenchymal stem cells. Biomaterials 2012, 33, 7686–7698. [Google Scholar] [CrossRef]
- Ker, E.D.; Nain, A.S.; Weiss, L.E.; Wang, J.; Suhan, J.; Amon, C.H.; Campbell, P.G. Bioprinting of growth factors onto aligned sub-micron fibrous scaffolds for simultaneous control of cell differentiation and alignment. Biomaterials 2011, 32, 8097–8107. [Google Scholar] [CrossRef]
- Lutolf, M.P.; Hubbell, J.A. Synthetic biomaterials as instructive extracellular microenvironments for morphogenesis in tissue engineering. Nat. Biotechnol. 2005, 23, 47–55. [Google Scholar] [CrossRef]
- Mizuno, M.; Fujisawa, R.; Kuboki, Y. Type I collagen-induced osteoblastic differentiation of bone-marrow cells mediated by collagen-α2β1 integrin interaction. J. Cell. Physiol. 2000, 184, 207–213. [Google Scholar] [CrossRef]
- Mizuno, M.; Kuboki, Y. Osteoblast-related gene expression of bone marrow cells during the osteoblastic differentiation induced by type I collagen. J. Biochem. 2001, 129, 133–138. [Google Scholar] [CrossRef] [PubMed]
- Teh, T.K.; Toh, S.-L.; Goh, J.C. Aligned hybrid silk scaffold for enhanced differentiation of mesenchymal stem cells into ligament fibroblasts. Tissue Eng. Part C Methods 2011, 17, 687–703. [Google Scholar] [CrossRef]
- Banes, A.J.; Lee, G.; Graff, R.; Otey, C.; Archambault, J.; Tsuzaki, M.; Elfervig, M.; Qi, J. Mechanical forces and signaling in connective tissue cells: Cellular mechanisms of detection, transduction, and responses to mechanical deformation. Curr. Opin. Orthop. 2001, 12, 389–396. [Google Scholar] [CrossRef]
- Hung, C.T.; Allen, F.D.; Pollack, S.R.; Attia, E.T.; Hannafin, J.A.; Torzilli, P.A. Intracellular calcium response of ACL and MCL ligament fibroblasts to fluid-induced shear stress. Cell. Signal. 1997, 9, 587–594. [Google Scholar] [CrossRef]
- Tsuzaki, M.; Bynum, D.; Almekinders, L.; Yang, X.; Faber, J.; Banes, A. ATP modulates load-inducible IL-1, COX 2, and MMP-3 gene expression in human tendon cells. J. Cell. Biochem. 2003, 89, 556–562. [Google Scholar] [CrossRef] [PubMed]
- Chiquet, M.; Gelman, L.; Lutz, R.; Maier, S. From mechanotransduction to extracellular matrix gene expression in fibroblasts. Biochim. Biophys. Acta (BBA) Mol. Cell Res. 2009, 1793, 911–920. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Barber, J.G.; Handorf, A.M.; Allee, T.J.; Li, W.-J. Braided nanofibrous scaffold for tendon and ligament tissue engineering. Tissue Eng. Part A 2013, 19, 1265–1274. [Google Scholar] [CrossRef]
- Li, W.-J.; Jiang, Y.J.; Tuan, R.S. Chondrocyte phenotype in engineered fibrous matrix is regulated by fiber size. Tissue Eng. 2006, 12, 1775–1785. [Google Scholar] [CrossRef]
- Ralphs, J.; Waggett, A.D.; Benjamin, M. Actin stress fibres and cell-cell adhesion molecules in tendons: Organisation in vivo and response to mechanical loading of tendon cells in vitro. Matrix Biol. 2002, 21, 67–74. [Google Scholar] [CrossRef]
- Diekman, B.O.; Christoforou, N.; Willard, V.P.; Sun, H.; Sanchez-Adams, J.; Leong, K.W.; Guilak, F. Cartilage tissue engineering using differentiated and purified induced pluripotent stem cells. Proc. Natl. Acad. Sci. USA 2012, 109, 19172–19177. [Google Scholar] [CrossRef] [Green Version]
- Li, W.-J.; Cooper, J.A.; Mauck, R.L.; Tuan, R.S. Fabrication and characterization of six electrospun poly(α-hydroxy ester)-based fibrous scaffolds for tissue engineering applications. Acta Biomater. 2006, 2, 377–385. [Google Scholar] [CrossRef] [PubMed]
- Rothrauff, B.B.; Lauro, B.B.; Yang, G.; Debski, R.E.; Musahl, V.; Tuan, R.S. Braided and stacked electrospun nanofibrous scaffolds for tendon and ligament tissue engineering. Tissue Eng. Part A 2017, 23, 378–389. [Google Scholar] [CrossRef] [PubMed]
- Orr, S.B.; Chainani, A.; Hippensteel, K.; Kishan, A.; Gilchrist, C.; Garrigues, N.W.; Ruch, D.S.; Guilak, F.; Little, D. Aligned multilayered electrospun scaffolds for rotator cuff tendon tissue engineering. Acta Biomater. 2015, 24, 117–126. [Google Scholar] [CrossRef] [Green Version]
- Vuornos, K.; Björninen, M.; Talvitie, E.; Paakinaho, K.; Kellomäki, M.; Huhtala, H.; Miettinen, S.; Seppänen-Kaijansinkko, R.; Haimi, S. Human adipose stem cells differentiated on braided polylactide scaffolds is a potential approach for tendon tissue engineering. Tissue Eng. Part A 2016, 22, 513–523. [Google Scholar] [CrossRef] [PubMed]
- Yin, Z.; Chen, X.; Song, H.-X.; Hu, J.-J.; Tang, Q.-M.; Zhu, T.; Shen, W.-L.; Chen, J.-L.; Liu, H.; Heng, B.C.; et al. Electrospun scaffolds for multiple tissues regeneration in vivo through topography dependent induction of lineage specific differentiation. Biomaterials 2015, 44, 173–185. [Google Scholar] [CrossRef]
- Liu, Y.; Zhou, G.; Cao, Y. Recent Progress in Cartilage Tissue Engineering—Our Experience and Future Directions. Engineering 2017, 3, 28–35. [Google Scholar] [CrossRef]
- Vinatier, C.; Guicheux, J. Cartilage tissue engineering: From biomaterials and stem cells to osteoarthritis treatments. Ann. Phys. Rehabil. Med. 2016, 59, 139–144. [Google Scholar] [CrossRef]
- Steadman, J.R.; Rodkey, W.G.; Rodrigo, J.J. Microfracture: Surgical Technique and Rehabilitation to Treat Chondral Defects. Clin. Orthop. Relat. Res. 2001, 391, S362–S369. [Google Scholar] [CrossRef]
- Hangody, L.; Füles, P. Autologous osteochondral mosaicplasty for the treatment of full-thickness defects of weight-bearing joints. J. Bone Jt. Surg. Am. Vol. 2003, 85, 25–32. [Google Scholar] [CrossRef]
- Brittberg, M.; Lindahl, A.; Nilsson, A.; Ohlsson, C.; Isaksson, O.; Peterson, L. Treatment of deep cartilage defects in the knee with autologous chondrocyte transplantation. New Engl. J. Med. 1994, 331, 889–895. [Google Scholar] [CrossRef]
- Spiller, K.L.; Maher, S.A.; Lowman, A.M. Hydrogels for the repair of articular cartilage defects. Tissue Eng. Part B Rev. 2011, 17, 281–299. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Moran, C.J.; Pascual-Garrido, C.; Chubinskaya, S.; Potter, H.G.; Warren, R.F.; Cole, B.J.; Rodeo, S.A. Restoration of articular cartilage. J. Bone Jt. Surg. Am. Vol. 2014, 96, 336–344. [Google Scholar] [CrossRef] [Green Version]
- Huey, D.J.; Hu, J.C.; Athanasiou, K.A. Unlike bone, cartilage regeneration remains elusive. Science 2012, 338, 917–921. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Van Osch, G.J.V.M.; Brittberg, M.; Dennis, J.; Bastiaansen-Jenniskens, Y.; Erben, R.; Konttinen, Y.; Luyten, F. Cartilage repair: Past and future-lessons for regenerative medicine. J. Cell. Mol. Med. 2009, 13, 792–810. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jorgensen, C.; Gordeladze, J.; Noel, D. Tissue engineering through autologous mesenchymal stem cells. Curr. Opin. Biotechnol. 2004, 15, 406–410. [Google Scholar] [CrossRef]
- Caplan, A.I. Review: Mesenchymal stem cells: Cell-based reconstructive therapy in orthopedics. Tissue Eng. 2005, 11, 1198–1211. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nerurkar, N.L.; Baker, B.M.; Sen, S.; Wible, E.E.; Elliott, D.M.; Mauck, R.L. Nanofibrous biologic laminates replicate the form and function of the annulus fibrosus. Nat. Mater. 2009, 8, 986–992. [Google Scholar] [CrossRef] [Green Version]
- Baker, B.M.; Mauck, R.L. The effect of nanofiber alignment on the maturation of engineered meniscus constructs. Biomaterials 2007, 28, 1967–1977. [Google Scholar] [CrossRef] [Green Version]
- Baker, B.M.; Nathan, A.S.; Gee, A.O.; Mauck, R.L. The influence of an aligned nanofibrous topography on human mesenchymal stem cell fibrochondrogenesis. Biomaterials 2010, 31, 6190–6200. [Google Scholar] [CrossRef] [Green Version]
- Wu, Y.; Yang, Z.; Law, J.B.K.; He, A.Y.; Abbas, A.A.; Denslin, V.; Kamarul, T.; Hui, J.; Lee, E.H. The Combined effect of substrate stiffness and surface topography on chondrogenic differentiation of mesenchymal stem cells. Tissue Eng. Part A 2017, 23, 43–54. [Google Scholar] [CrossRef]
- Jurvelin, J.; Müller, D.J.; Wong, M.; Studer, D.; Engel, A.; Hunziker, E. Surface and subsurface morphology of bovine humeral articular cartilage as assessed by atomic force and transmission electron microscopy. J. Struct. Biol. 1996, 117, 45–54. [Google Scholar] [CrossRef] [PubMed]
- Wu, Y.-N.; Yang, Z.; Hui, J.H.; Ouyang, H.-W.; Lee, E.H. Cartilaginous ECM component-modification of the micro-bead culture system for chondrogenic differentiation of mesenchymal stem cells. Biomaterials 2007, 28, 4056–4067. [Google Scholar] [CrossRef] [PubMed]
- Varghese, S.; Hwang, N.S.; Canver, A.C.; Theprungsirikul, P.; Lin, D.W.; Elisseeff, J. Chondroitin sulfate based niches for chondrogenic differentiation of mesenchymal stem cells. Matrix Biol. 2008, 27, 12–21. [Google Scholar] [CrossRef] [PubMed]
- Klein, T.J.; Malda, J.; Sah, R.L.; Hutmacher, D.W. Tissue engineering of articular cartilage with biomimetic zones. Tissue Eng. Part B Rev. 2009, 15, 143–157. [Google Scholar] [CrossRef]
- Hayes, A.J.; Hall, A.; Brown, L.; Tubo, R.; Caterson, B. Macromolecular organization and in vitro growth characteristics of scaffold-free neocartilage grafts. J. Histochem. Cytochem. 2007, 55, 853–866. [Google Scholar] [CrossRef] [Green Version]
- McMahon, R.E.; Wang, L.; Skoracki, R.; Mathur, A.B. Development of nanomaterials for bone repair and regeneration. J. Biomed. Mater. Res. Part B Appl. Biomater. 2012, 101, 387–397. [Google Scholar] [CrossRef]
- Walpole, A.R.; Xia, Z.; Wilson, C.W.; Triffitt, J.T.; Wilshaw, P.R. A novel nano-porous alumina biomaterial with potential for loading with bioactive materials. J. Biomed. Mater. Res. Part A 2009, 90, 46–54. [Google Scholar] [CrossRef]
- Zhang, B.; Pei, X.; Song, P.; Sun, H.; Li, H.; Fan, Y.; Jiang, Q.; Zhou, C.; Zhang, X. Porous bioceramics produced by inkjet 3D printing: Effect of printing ink formulation on the ceramic macro and micro porous architectures control. Compos. Part B Eng. 2018, 155, 112–121. [Google Scholar] [CrossRef]
- Schumacher, M.; Deisinger, U.; Detsch, R.; Ziegler, G. Indirect rapid prototyping of biphasic calcium phosphate scaffolds as bone substitutes: Influence of phase composition, macroporosity and pore geometry on mechanical properties. J. Mater. Sci. Mater. Electron. 2010, 21, 3119–3127. [Google Scholar] [CrossRef]
- Wu, C.; Ramaswamy, Y.; Zreiqat, H. Porous diopside (CaMgSi2O6) scaffold: A promising bioactive material for bone tissue engineering. Acta Biomater. 2010, 6, 2237–2245. [Google Scholar] [CrossRef]
- Pei, X.; Ma, L.; Zhang, B.; Sun, J.; Sun, Y.; Fan, Y.; Gou, Z.; Zhou, C.; Zhang, X. Creating hierarchical porosity hydroxyapatite scaffolds with osteoinduction by three-dimensional printing and microwave sintering. Biofabrication 2017, 9, 045008. [Google Scholar] [CrossRef]
- Daugela, P.; Pranskunas, M.; Juodzbalys, G.; Liesiene, J.; Baniukaitiene, O.; Afonso, A.; Gomes, P.S. Novel cellulose/hydroxyapatite scaffolds for bone tissue regeneration: In vitro and in vivo study. J. Tissue Eng. Regen. Med. 2018, 12, 1195–1208. [Google Scholar] [CrossRef]
- Godoy-Gallardo, M.; Portolés-Gil, N.; López-Periago, A.M.; Domingo, C.; Hosta-Rigau, L. Multi-layered polydopamine coatings for the immobilization of growth factors onto highly-interconnected and bimodal PCL/HA-based scaffolds. Mater. Sci. Eng. C 2020, 117, 111245. [Google Scholar] [CrossRef] [PubMed]
- Godoy-Gallardo, M.; Portolés-Gil, N.; López-Periago, A.M.; Domingo, C.; Hosta-Rigau, L. Immobilization of BMP-2 and VEGF within multilayered polydopamine-coated scaffolds and the resulting osteogenic and angiogenic synergy of co-cultured human mesenchymal stem cells and human endothelial progenitor cells. Int. J. Mol. Sci. 2020, 21, 6418. [Google Scholar] [CrossRef] [PubMed]
- Zhang, K.; Fan, Y.; Dunne, N.; Li, X. Effect of microporosity on scaffolds for bone tissue engineering. Regen. Biomater. 2018, 5, 115–124. [Google Scholar] [CrossRef] [PubMed]
- Lord, M.S.; Foss, M.; Besenbacher, F. Influence of nanoscale surface topography on protein adsorption and cellular response. Nano Today 2010, 5, 66–78. [Google Scholar] [CrossRef]
- Zhao, F.; Mc Garrigle, M.J.; Vaughan, T.J.; McNamara, L.M. In silico study of bone tissue regeneration in an idealised porous hydrogel scaffold using a mechano-regulation algorithm. Biomech. Model. Mechanobiol. 2017, 17, 5–18. [Google Scholar] [CrossRef]
- Raafat, A.I.; Abd-Allah, W.M. In vitro apatite forming ability and ketoprofen release of radiation synthesized (gelatin-polyvinyl alcohol)/bioglass composite scaffolds for bone tissue regeneration. Polym. Compos. 2016, 39, 606–615. [Google Scholar] [CrossRef]
- Shim, K.-S.; Kim, H.J.; Yun, Y.-P.; Jeon, D.I.; Kim, H.J.; Park, K.; Song, H.-R. Surface immobilization of biphasic calcium phosphate nanoparticles on 3D printed poly (caprolactone) scaffolds enhances osteogenesis and bone tissue regeneration. J. Ind. Eng. Chem. 2017, 55, 101–109. [Google Scholar] [CrossRef]
- Bružauskaitė, I.; Bironaitė, D.; Bagdonas, E.; Bernotienė, E. Scaffolds and cells for tissue regeneration: Different scaffold pore sizes-different cell effects. Cytotechnology 2015, 68, 355–369. [Google Scholar] [CrossRef] [Green Version]
- Hulbert, S.F.; Young, F.A.; Mathews, R.S.; Klawitter, J.J.; Talbert, C.D.; Stelling, F.H. Potential of ceramic materials as permanently implantable skeletal prostheses. J. Biomed. Mater. Res. 1970, 4, 433–456. [Google Scholar] [CrossRef] [PubMed]
- Ashman, A.; Moss, M.L. Implantation of porous polymethylmethacrylate resin for tooth and bone replacement. J. Prosthet. Dent. 1977, 37, 657–665. [Google Scholar] [CrossRef]
- Murphy, C.M.; Haugh, M.G.; O’Brien, F.J. The effect of mean pore size on cell attachment, proliferation and migration in collagen–glycosaminoglycan scaffolds for bone tissue engineering. Biomaterials 2010, 31, 461–466. [Google Scholar] [CrossRef] [PubMed]
- Nehrer, S.; Breinan, H.A.; Ramappa, A.; Young, G.; Shortkroff, S.; Louie, L.K.; Sledge, C.B.; Yannas, I.V.; Spector, M. Matrix collagen type and pore size influence behaviour of seeded canine chondrocytes. Biomaterials 1997, 18, 769–776. [Google Scholar] [CrossRef]
- Lee, S.J.; Lee, I.W.; Lee, Y.M.; Lee, H.B.; Khang, G. Macroporous biodegradable natural/synthetic hybrid scaffolds as small intestine submucosa impregnated poly(D,L-lactide-co-glycolide) for tissue-engineered bone. J. Biomater. Sci. Polym. Ed. 2004, 15, 1003–1017. [Google Scholar] [CrossRef] [PubMed]
- Baksh, D.; Davies, J.E.; Kim, S. Three-dimensional matrices of calcium polyphosphates support bone growth in vitro and in vivo. J. Mater. Sci. Mater. Electron. 1998, 9, 743–748. [Google Scholar] [CrossRef] [PubMed]
- Peter, S.J.; Miller, M.J.; Yasko, A.W.; Yaszemski, M.J.; Mikos, A.G. Polymer concepts in tissue engineering. J. Biomed. Mater. Res. 1998, 43. [Google Scholar] [CrossRef]
- Ishaug, S.L.; Crane, G.M.; Miller, M.J.; Yasko, A.W.; Yaszemski, M.J.; Mikos, A.G. Bone formation by three-dimensional stromal osteoblast culture in biodegradable polymer scaffolds. J. Biomed. Mater. Res. 1997, 36. [Google Scholar] [CrossRef]
- Akay, G.; Birch, M.; Bokhari, M. Microcellular polyHIPE polymer supports osteoblast growth and bone formation in vitro. Biomaterials 2004, 25, 3991–4000. [Google Scholar] [CrossRef]
- Kuboki, Y.; Jin, Q.; Takita, H. Delivery Systems for the BMPs Geometry of carriers controlling phenotypic expression in BMP-induced osteogenesis and chondrogenesis. BMPS 2001, 83, 105–115. [Google Scholar]
- Van Tienen, T.G.; Heijkants, R.G.J.C.; De Groot, J.H.; Pennings, A.J.; Schouten, A.J.; Veth, R.P.H.; Buma, P. Replacement of the Knee Meniscus by a Porous Polymer Implant. Am. J. Sports Med. 2006, 34, 64–71. [Google Scholar] [CrossRef] [PubMed]
- Matsiko, A.; Gleeson, J.P.; O’Brien, F.J. Scaffold mean pore size influences mesenchymal stem cell chondrogenic differentiation and matrix deposition. Tissue Eng. Part A 2015, 21, 486–497. [Google Scholar] [CrossRef] [PubMed]
- Sha’Ban, M.; Kim, S.H.; Ruszymah, B.; Khang, G. Fibrin and poly(lactic-co-glycolic acid) hybrid scaffold promotes early chondrogenesis of articular chondrocytes: An in vitro study. J. Orthop. Surg. Res. 2008, 3, 17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, J.; Nie, H.; Xu, Z.; Niu, X.; Guo, S.; Yin, J.; Guo, F.; Li, G.; Wang, Y.; Zhang, C. The effect of 3D nanofibrous scaffolds on the chondrogenesis of induced pluripotent stem cells and their application in restoration of cartilage defects. PLoS ONE 2014, 9, e111566. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Long, M.; Rack, H. Titanium alloys in total joint replacement-A materials science perspective. Biomaterials 1998, 19, 1621–1639. [Google Scholar] [CrossRef]
- Niinomi, M. Recent research and development in titanium alloys for biomedical applications and healthcare goods. Sci. Technol. Adv. Mater. 2003, 4, 445–454. [Google Scholar] [CrossRef] [Green Version]
- Liu, X.; Chu, P.K.; Ding, C. Surface modification of titanium, titanium alloys, and related materials for biomedical applications. Mater. Sci. Eng. R Rep. 2004, 47, 49–121. [Google Scholar] [CrossRef] [Green Version]
- Wang, X.; Xu, S.; Zhou, S.; Xu, W.; Leary, M.; Choong, P.; Qian, M.; Brandt, M.; Xie, Y.M. Topological design and additive manufacturing of porous metals for bone scaffolds and orthopaedic implants: A review. Biomaterials 2016, 83, 127–141. [Google Scholar] [CrossRef]
- Itälä, A.I.; Ylänen, H.O.; Ekholm, C.; Karlsson, K.H.; Aro, H.T. Pore diameter of more than 100 μm is not requisite for bone ingrowth in rabbits. J. Biomed. Mater. Res. 2001, 58, 679–683. [Google Scholar] [CrossRef]
- Braem, A.; Chaudhari, A.; Cardoso, M.V.; Schrooten, J.; Duyck, J.; Vleugels, J. Peri- and intra-implant bone response to microporous Ti coatings with surface modification. Acta Biomater. 2014, 10, 986–995. [Google Scholar] [CrossRef] [Green Version]
- Taniguchi, N.; Fujibayashi, S.; Takemoto, M.; Sasaki, K.; Otsuki, B.; Nakamura, T.; Matsushita, T.; Kokubo, T.; Matsuda, S. Effect of pore size on bone ingrowth into porous titanium implants fabricated by additive manufacturing: An in vivo experiment. Mater. Sci. Eng. C 2016, 59, 690–701. [Google Scholar] [CrossRef] [Green Version]
- Chan, O.; Coathup, M.; Nesbitt, A.; Ho, C.-Y.; Hing, K.; Buckland, T.; Campion, C.; Blunn, G.W. The effects of microporosity on osteoinduction of calcium phosphate bone graft substitute biomaterials. Acta Biomater. 2012, 8, 2788–2794. [Google Scholar] [CrossRef] [PubMed]
- Campion, C.R.; Chander, C.; Buckland, T.; Hing, K. Increasing strut porosity in silicate-substituted calcium-phosphate bone graft substitutes enhances osteogenesis. J. Biomed. Mater. Res. Part B Appl. Biomater. 2011, 97, 245–254. [Google Scholar] [CrossRef] [PubMed]
- Aarvold, A.; Smith, J.O.; Tayton, E.R.; Lanham, S.; Chaudhuri, J.B.; Turner, I.G.; Oreffo, R.O. The effect of porosity of a biphasic ceramic scaffold on human skeletal stem cell growth and differentiationin vivo. J. Biomed. Mater. Res. Part A 2013, 101, 3431–3437. [Google Scholar] [CrossRef]
- Gibson, L.J.; Ashby, M.F. Cellular Solids: Structure and Properties; Cambridge University Press: Cambridge, UK, 1999. [Google Scholar]
- Story, B.J.; Wagner, W.R.; Gaisser, D.M.; Cook, S.D.; Rust-Dawicki, A.M. In vivo performance of a modified CSTi dental implant coating. Int. J. Oral Maxillofac. Implant. 1998, 13, 749–757. [Google Scholar]
- Eqtesadi, S.; Motealleh, A.; Pajares, A.; Miranda, P. Effect of milling media on processing and performance of 13-93 bioactive glass scaffolds fabricated by robocasting. Ceram. Int. 2015, 41, 1379–1389. [Google Scholar] [CrossRef]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2020 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Cun, X.; Hosta-Rigau, L. Topography: A Biophysical Approach to Direct the Fate of Mesenchymal Stem Cells in Tissue Engineering Applications. Nanomaterials 2020, 10, 2070. https://doi.org/10.3390/nano10102070
Cun X, Hosta-Rigau L. Topography: A Biophysical Approach to Direct the Fate of Mesenchymal Stem Cells in Tissue Engineering Applications. Nanomaterials. 2020; 10(10):2070. https://doi.org/10.3390/nano10102070
Chicago/Turabian StyleCun, Xingli, and Leticia Hosta-Rigau. 2020. "Topography: A Biophysical Approach to Direct the Fate of Mesenchymal Stem Cells in Tissue Engineering Applications" Nanomaterials 10, no. 10: 2070. https://doi.org/10.3390/nano10102070
APA StyleCun, X., & Hosta-Rigau, L. (2020). Topography: A Biophysical Approach to Direct the Fate of Mesenchymal Stem Cells in Tissue Engineering Applications. Nanomaterials, 10(10), 2070. https://doi.org/10.3390/nano10102070