Skip to main content
Top
Published in: Journal of Orthopaedic Surgery and Research 1/2016

Open Access 01-12-2016 | Review

Matrix scaffolding for stem cell guidance toward skeletal muscle tissue engineering

Authors: Claudia Fuoco, Lucia Lisa Petrilli, Stefano Cannata, Cesare Gargioli

Published in: Journal of Orthopaedic Surgery and Research | Issue 1/2016

Login to get access

Abstract

Extracellular matrix (ECM) is composed of many types of fibrous structural proteins and glycosaminoglycans. This important cell component not only provides a support for cells but is also actively involved in cell-cell interaction, proliferation, migration, and differentiation, representing, therefore, no longer only a mere static structural scaffold for cells but rather a dynamic and versatile compartment. This aspect leads to the need for investigating new bio-inspired scaffolds or biomaterials, able to mimic ECM in tissue engineering. This new field of research finds particular employment in skeletal muscle tissue regeneration, due to the inability of this complex tissue to recover volumetric muscle loss (VML), after severe injury. Usually, this is the result of traumatic incidents, tumor ablations, or pathological states that lead to the destruction of a large amount of tissue, including connective tissue and basement membrane. Therefore, skeletal muscle tissue engineering represents a valid alternative to overcome this problem.
Here, we described a series of natural and synthetic biomaterials employed as ECM mimics for their ability to recreate the correct muscle stem cell niche, by promoting myogenic stem cell differentiation and so, positively affecting muscle repair.
Literature
2.
go back to reference Arnold L, Henry A, Poron F, Baba-Amer Y, van Rooijen N, Plonquet A, et al. Inflammatory monocytes recruited after skeletal muscle injury switch into antiinflammatory macrophages to support myogenesis. J Exp Med. 2007;204:1057–69. Rockefeller Univ Press.CrossRefPubMedPubMedCentral Arnold L, Henry A, Poron F, Baba-Amer Y, van Rooijen N, Plonquet A, et al. Inflammatory monocytes recruited after skeletal muscle injury switch into antiinflammatory macrophages to support myogenesis. J Exp Med. 2007;204:1057–69. Rockefeller Univ Press.CrossRefPubMedPubMedCentral
3.
go back to reference Relaix F, Zammit PS. Satellite cells are essential for skeletal muscle regeneration: the cell on the edge returns centre stage. Development. 2012;139:2845–56.CrossRefPubMed Relaix F, Zammit PS. Satellite cells are essential for skeletal muscle regeneration: the cell on the edge returns centre stage. Development. 2012;139:2845–56.CrossRefPubMed
4.
go back to reference Brack AS, Rando TA. Tissue-specific stem cells: lessons from the skeletal muscle satellite cell. Stem Cell. 2012;10:504–14. Elsevier Inc. Brack AS, Rando TA. Tissue-specific stem cells: lessons from the skeletal muscle satellite cell. Stem Cell. 2012;10:504–14. Elsevier Inc.
5.
go back to reference Chargé SBP, Rudnicki MA. Cellular and molecular regulation of muscle regeneration. Physiol Rev. 2004;84:209–38. American Physiological Society.CrossRefPubMed Chargé SBP, Rudnicki MA. Cellular and molecular regulation of muscle regeneration. Physiol Rev. 2004;84:209–38. American Physiological Society.CrossRefPubMed
6.
go back to reference Wang YX, Rudnicki MA. Satellite cells, the engines of muscle repair. Nat Rev Mol Cell Biol. 2012;13:127–33. Wang YX, Rudnicki MA. Satellite cells, the engines of muscle repair. Nat Rev Mol Cell Biol. 2012;13:127–33.
7.
8.
go back to reference Theocharis AD, Skandalis SS, Gialeli C, Karamanos NK. Extracellular matrix structure. Adv Drug Deliv Rev. 2015;1–77. Elsevier B.V. Theocharis AD, Skandalis SS, Gialeli C, Karamanos NK. Extracellular matrix structure. Adv Drug Deliv Rev. 2015;1–77. Elsevier B.V.
10.
go back to reference Kadler KE, Baldock C, Bella J, Boot-Handford RP. Collagens at a glance. J Cell Sci. 2007;120:1955–8. The Company of Biologists Ltd.CrossRefPubMed Kadler KE, Baldock C, Bella J, Boot-Handford RP. Collagens at a glance. J Cell Sci. 2007;120:1955–8. The Company of Biologists Ltd.CrossRefPubMed
11.
go back to reference Hoshiba T, Chen G, Endo C, Maruyama H, Wakui M, Nemoto E, et al. Decellularized extracellular matrix as an in vitro model to study the comprehensive roles of the ECM in stem cell differentiation. Stem Cells Int. 2016;2016:1–10.CrossRef Hoshiba T, Chen G, Endo C, Maruyama H, Wakui M, Nemoto E, et al. Decellularized extracellular matrix as an in vitro model to study the comprehensive roles of the ECM in stem cell differentiation. Stem Cells Int. 2016;2016:1–10.CrossRef
12.
go back to reference Engler AJ, Sen S, Sweeney HL, Discher DE. Matrix elasticity directs stem cell lineage specification. Cell. 2006;126:677–89.CrossRefPubMed Engler AJ, Sen S, Sweeney HL, Discher DE. Matrix elasticity directs stem cell lineage specification. Cell. 2006;126:677–89.CrossRefPubMed
13.
go back to reference Nagaprashantha LD, Vatsyayan R, Lelsani PCR, Awasthi S, Singhal SS. The sensors and regulators of cell-matrix surveillance in anoikis resistance of tumors. Int J Cancer. 2011;128:743–52. Wiley Subscription Services, Inc., A Wiley Company.CrossRefPubMed Nagaprashantha LD, Vatsyayan R, Lelsani PCR, Awasthi S, Singhal SS. The sensors and regulators of cell-matrix surveillance in anoikis resistance of tumors. Int J Cancer. 2011;128:743–52. Wiley Subscription Services, Inc., A Wiley Company.CrossRefPubMed
14.
go back to reference Cox TR, Erler JT. Remodeling and homeostasis of the extracellular matrix: implications for fibrotic diseases and cancer. Dis Model Mech. 2011;4:165–78.CrossRefPubMedPubMedCentral Cox TR, Erler JT. Remodeling and homeostasis of the extracellular matrix: implications for fibrotic diseases and cancer. Dis Model Mech. 2011;4:165–78.CrossRefPubMedPubMedCentral
15.
go back to reference Rozario T, DeSimone DW. The extracellular matrix in development and morphogenesis: a dynamic view. Dev Biol. 2010;341:126–40.CrossRefPubMed Rozario T, DeSimone DW. The extracellular matrix in development and morphogenesis: a dynamic view. Dev Biol. 2010;341:126–40.CrossRefPubMed
16.
go back to reference Heino J. The collagen family members as cell adhesion proteins. Bioessays. 2007;29:1001–10. Wiley Subscription Services, Inc., A Wiley Company.CrossRefPubMed Heino J. The collagen family members as cell adhesion proteins. Bioessays. 2007;29:1001–10. Wiley Subscription Services, Inc., A Wiley Company.CrossRefPubMed
17.
go back to reference De Wever O, Demetter P, Mareel M, Bracke M. Stromal myofibroblasts are drivers of invasive cancer growth. Int J Cancer. 2008;123:2229–38. Wiley Subscription Services, Inc., A Wiley Company.CrossRefPubMed De Wever O, Demetter P, Mareel M, Bracke M. Stromal myofibroblasts are drivers of invasive cancer growth. Int J Cancer. 2008;123:2229–38. Wiley Subscription Services, Inc., A Wiley Company.CrossRefPubMed
18.
go back to reference Tsang KY, Cheung MCH, Chan D, Cheah KSE. The developmental roles of the extracellular matrix: beyond structure to regulation. Cell Tissue Res. 2010;339:93–110. Springer-Verlag.CrossRefPubMed Tsang KY, Cheung MCH, Chan D, Cheah KSE. The developmental roles of the extracellular matrix: beyond structure to regulation. Cell Tissue Res. 2010;339:93–110. Springer-Verlag.CrossRefPubMed
19.
go back to reference Aumailley M, Bruckner-Tuderman L, Carter WG, Deutzmann R, Edgar D, Ekblom P, et al. A simplified laminin nomenclature. Matrix Biol. 2005;24:326–32.CrossRefPubMed Aumailley M, Bruckner-Tuderman L, Carter WG, Deutzmann R, Edgar D, Ekblom P, et al. A simplified laminin nomenclature. Matrix Biol. 2005;24:326–32.CrossRefPubMed
20.
go back to reference Theocharis AD, Skandalis SS, Tzanakakis GN, Karamanos NK. Proteoglycans in health and disease: novel roles for proteoglycans in malignancy and their pharmacological targeting. FEBS Journal. 2010;277:3904–23.CrossRefPubMed Theocharis AD, Skandalis SS, Tzanakakis GN, Karamanos NK. Proteoglycans in health and disease: novel roles for proteoglycans in malignancy and their pharmacological targeting. FEBS Journal. 2010;277:3904–23.CrossRefPubMed
23.
go back to reference Jones DL, Wagers AJ. No place like home: anatomy and function of the stem cell niche. Nat Rev Mol Cell Biol. 2008;9:11–21.CrossRefPubMed Jones DL, Wagers AJ. No place like home: anatomy and function of the stem cell niche. Nat Rev Mol Cell Biol. 2008;9:11–21.CrossRefPubMed
24.
go back to reference Teodori L, Costa A, Marzio R, Perniconi B, Coletti D, Adamo S, et al. Native extracellular matrix: a new scaffolding platform for repair of damaged muscle. Front Physiol. 2014;5:590–10.CrossRef Teodori L, Costa A, Marzio R, Perniconi B, Coletti D, Adamo S, et al. Native extracellular matrix: a new scaffolding platform for repair of damaged muscle. Front Physiol. 2014;5:590–10.CrossRef
25.
go back to reference Grasman JM, Zayas MJ, Page R, Pins GD. Biomimetic scaffolds for regeneration of volumetric muscle loss in skeletal muscle injuries. Acta Biomaterialia. 2015;1–49. Acta Materialia Inc. Grasman JM, Zayas MJ, Page R, Pins GD. Biomimetic scaffolds for regeneration of volumetric muscle loss in skeletal muscle injuries. Acta Biomaterialia. 2015;1–49. Acta Materialia Inc.
26.
27.
go back to reference Cezar CA, Mooney DJ. Biomaterial-based delivery for skeletal muscle repair. Adv Drug Deliv Rev. 2014;1–44. Elsevier B.V. Cezar CA, Mooney DJ. Biomaterial-based delivery for skeletal muscle repair. Adv Drug Deliv Rev. 2014;1–44. Elsevier B.V.
28.
go back to reference Mantero S. Skeletal muscle tissue engineering: strategies for volumetric constructs. 2014. p. 1–13. Mantero S. Skeletal muscle tissue engineering: strategies for volumetric constructs. 2014. p. 1–13.
29.
go back to reference Grogan BF, Hsu JR, Consortium STR. Volumetric muscle loss. J Am Acad Orthop Surg. 2011;19 Suppl 1:S35–7.CrossRefPubMed Grogan BF, Hsu JR, Consortium STR. Volumetric muscle loss. J Am Acad Orthop Surg. 2011;19 Suppl 1:S35–7.CrossRefPubMed
30.
go back to reference Qazi TH, Mooney DJ, Pumberger M, Geissler S, Duda GN. Biomaterials based strategies for skeletal muscle tissue engineering: existing technologies and future trends. Biomaterials. 2015;53:502–21.CrossRefPubMed Qazi TH, Mooney DJ, Pumberger M, Geissler S, Duda GN. Biomaterials based strategies for skeletal muscle tissue engineering: existing technologies and future trends. Biomaterials. 2015;53:502–21.CrossRefPubMed
31.
go back to reference Hurd SA, Bhatti NM, Walker AM, Kasukonis BM, Wolchok JC. Development of a biological scaffold engineered using the extracellular matrix secreted by skeletal muscle cells. Biomaterials. 2015;49:9–17.CrossRefPubMedPubMedCentral Hurd SA, Bhatti NM, Walker AM, Kasukonis BM, Wolchok JC. Development of a biological scaffold engineered using the extracellular matrix secreted by skeletal muscle cells. Biomaterials. 2015;49:9–17.CrossRefPubMedPubMedCentral
32.
go back to reference Shandalov Y, Egozi D, Freiman A, Rosenfeld D, Levenberg S. A method for constructing vascularized muscle flap. Methods. 2015;84:70–5.CrossRefPubMed Shandalov Y, Egozi D, Freiman A, Rosenfeld D, Levenberg S. A method for constructing vascularized muscle flap. Methods. 2015;84:70–5.CrossRefPubMed
33.
go back to reference Merritt EK, Hammers DW, Tierney M, Suggs LJ, Walters TJ, Farrar RP. Functional assessment of skeletal muscle regeneration utilizing homologous extracellular matrix as scaffolding. Tissue Eng Part A. 2010;16:1395–405.CrossRefPubMed Merritt EK, Hammers DW, Tierney M, Suggs LJ, Walters TJ, Farrar RP. Functional assessment of skeletal muscle regeneration utilizing homologous extracellular matrix as scaffolding. Tissue Eng Part A. 2010;16:1395–405.CrossRefPubMed
34.
go back to reference Karbalaeikhani A, Saied A, Heshmati A. Effectiveness of the gastrocsoleous flap for coverage of soft tissue defects in leg with emphasis on the distal third. Arch Bone Jt Surg. 2015;3:193–7.PubMedPubMedCentral Karbalaeikhani A, Saied A, Heshmati A. Effectiveness of the gastrocsoleous flap for coverage of soft tissue defects in leg with emphasis on the distal third. Arch Bone Jt Surg. 2015;3:193–7.PubMedPubMedCentral
35.
go back to reference Stern-Straeter J, Riedel F, Bran G, Hörmann K, Goessler UR. Advances in skeletal muscle tissue engineering. In Vivo. 2007;21:435–44.PubMed Stern-Straeter J, Riedel F, Bran G, Hörmann K, Goessler UR. Advances in skeletal muscle tissue engineering. In Vivo. 2007;21:435–44.PubMed
36.
go back to reference Fuoco C, Cannata S, Gargioli C. Could a functional artificial skeletal muscle be useful in muscle wasting? Curr Opin Clin Nutr Metab Care. 2016;1–6. Fuoco C, Cannata S, Gargioli C. Could a functional artificial skeletal muscle be useful in muscle wasting? Curr Opin Clin Nutr Metab Care. 2016;1–6.
37.
go back to reference McCullen SD, Chow AGY, Stevens MM. In vivo tissue engineering of musculoskeletal tissues. Curr Opin Biotechnol. 2011;22:715–20.CrossRefPubMed McCullen SD, Chow AGY, Stevens MM. In vivo tissue engineering of musculoskeletal tissues. Curr Opin Biotechnol. 2011;22:715–20.CrossRefPubMed
38.
go back to reference Lü S-H, Wang H-B, Liu H, Wang H-P, Lin Q-X, Li D-X, et al. Reconstruction of engineered uterine tissues containing smooth muscle layer in collagen/matrigel scaffold in vitro. Tissue Eng Part A. 2009;15:1611–8. Mary Ann Liebert, Inc. 140 Huguenot Street, 3rd Floor New Rochelle, NY 10801 USA.CrossRefPubMed Lü S-H, Wang H-B, Liu H, Wang H-P, Lin Q-X, Li D-X, et al. Reconstruction of engineered uterine tissues containing smooth muscle layer in collagen/matrigel scaffold in vitro. Tissue Eng Part A. 2009;15:1611–8. Mary Ann Liebert, Inc. 140 Huguenot Street, 3rd Floor New Rochelle, NY 10801 USA.CrossRefPubMed
39.
go back to reference van Wachem PB, van Luyn MJ, da Costa ML. Myoblast seeding in a collagen matrix evaluated in vitro. J Biomed Mater Res. 1996;30:353–60.CrossRefPubMed van Wachem PB, van Luyn MJ, da Costa ML. Myoblast seeding in a collagen matrix evaluated in vitro. J Biomed Mater Res. 1996;30:353–60.CrossRefPubMed
40.
go back to reference Karabekmez FE, Duymaz A, Moran SL. Early clinical outcomes with the use of decellularized nerve allograft for repair of sensory defects within the hand. Hand (N Y). 2009;4:245–9. SAGE Publications.CrossRef Karabekmez FE, Duymaz A, Moran SL. Early clinical outcomes with the use of decellularized nerve allograft for repair of sensory defects within the hand. Hand (N Y). 2009;4:245–9. SAGE Publications.CrossRef
41.
go back to reference Qing Q, Qin T. Optimal method for rat skeletal muscle decellularization. Zhongguo Xiu Fu Chong Jian Wai Ke Za Zhi. 2009;23:836–9.PubMed Qing Q, Qin T. Optimal method for rat skeletal muscle decellularization. Zhongguo Xiu Fu Chong Jian Wai Ke Za Zhi. 2009;23:836–9.PubMed
42.
go back to reference Gillies AR, Smith LR, Lieber RL, Varghese S. Method for decellularizing skeletal muscle without detergents or proteolytic enzymes. Tissue Eng Part C Methods. 2011;17:383–9.CrossRefPubMed Gillies AR, Smith LR, Lieber RL, Varghese S. Method for decellularizing skeletal muscle without detergents or proteolytic enzymes. Tissue Eng Part C Methods. 2011;17:383–9.CrossRefPubMed
43.
go back to reference Lin C-H, Yang J-R, Chiang N-J, Ma H, Tsay R-Y. Evaluation of decellularized extracellular matrix of skeletal muscle for tissue engineering. Int J Artif Organs. 2014;37:546–55.CrossRefPubMed Lin C-H, Yang J-R, Chiang N-J, Ma H, Tsay R-Y. Evaluation of decellularized extracellular matrix of skeletal muscle for tissue engineering. Int J Artif Organs. 2014;37:546–55.CrossRefPubMed
44.
go back to reference Zhang J, Hu ZQ, Turner NJ, Teng SF, Cheng WY, Zhou HY, et al. Perfusion-decellularized skeletal muscle as a three-dimensional scaffold with a vascular network template. Biomaterials. 2016;89:114–26.CrossRefPubMed Zhang J, Hu ZQ, Turner NJ, Teng SF, Cheng WY, Zhou HY, et al. Perfusion-decellularized skeletal muscle as a three-dimensional scaffold with a vascular network template. Biomaterials. 2016;89:114–26.CrossRefPubMed
46.
go back to reference Perniconi B, Costa A, Aulino P, Teodori L, Adamo S, Coletti D. The pro-myogenic environment provided by whole organ scale acellular scaffolds from skeletal muscle. Biomaterials. 2011;32:7870–82.CrossRefPubMed Perniconi B, Costa A, Aulino P, Teodori L, Adamo S, Coletti D. The pro-myogenic environment provided by whole organ scale acellular scaffolds from skeletal muscle. Biomaterials. 2011;32:7870–82.CrossRefPubMed
47.
go back to reference De Coppi P, Bellini S, Conconi MT, Sabatti M, Simonato E, Gamba PG, et al. Myoblast-acellular skeletal muscle matrix constructs guarantee a long-term repair of experimental full-thickness abdominal wall defects. Tissue Eng. 2006;12:1929–36. Mary Ann Liebert, Inc. 2 Madison Avenue Larchmont, NY 10538 USA.CrossRefPubMed De Coppi P, Bellini S, Conconi MT, Sabatti M, Simonato E, Gamba PG, et al. Myoblast-acellular skeletal muscle matrix constructs guarantee a long-term repair of experimental full-thickness abdominal wall defects. Tissue Eng. 2006;12:1929–36. Mary Ann Liebert, Inc. 2 Madison Avenue Larchmont, NY 10538 USA.CrossRefPubMed
50.
go back to reference Badylak SF. Decellularized allogeneic and xenogeneic tissue as a bioscaffold for regenerative medicine: factors that influence the host response. Ann Biomed Eng. 2014;42:1517–27. Springer US.CrossRefPubMed Badylak SF. Decellularized allogeneic and xenogeneic tissue as a bioscaffold for regenerative medicine: factors that influence the host response. Ann Biomed Eng. 2014;42:1517–27. Springer US.CrossRefPubMed
51.
go back to reference Wolf MT, Dearth CL, Sonnenberg SB, Loboa EG, Badylak SF. Naturally derived and synthetic scaffolds for skeletal muscle reconstruction. Adv Drug Deliv Rev. 2014;1–14. Elsevier B.V. Wolf MT, Dearth CL, Sonnenberg SB, Loboa EG, Badylak SF. Naturally derived and synthetic scaffolds for skeletal muscle reconstruction. Adv Drug Deliv Rev. 2014;1–14. Elsevier B.V.
52.
go back to reference Choi JS, Kim JD, Yoon HS, Cho YW. Full-thickness skin wound healing using human placenta-derived extracellular matrix containing bioactive molecules. Tissue Eng Part A. 2013;19:329–39.CrossRefPubMed Choi JS, Kim JD, Yoon HS, Cho YW. Full-thickness skin wound healing using human placenta-derived extracellular matrix containing bioactive molecules. Tissue Eng Part A. 2013;19:329–39.CrossRefPubMed
53.
go back to reference Crapo PM, Medberry CJ, Reing JE, Tottey S, van der Merwe Y, Jones KE, et al. Biologic scaffolds composed of central nervous system extracellular matrix. Biomaterials. 2012;33:3539–47. Elsevier Ltd.CrossRefPubMedPubMedCentral Crapo PM, Medberry CJ, Reing JE, Tottey S, van der Merwe Y, Jones KE, et al. Biologic scaffolds composed of central nervous system extracellular matrix. Biomaterials. 2012;33:3539–47. Elsevier Ltd.CrossRefPubMedPubMedCentral
54.
go back to reference Hoganson DM, Owens GE, O'Doherty EM, Bowley CM, Goldman SM, Harilal DO, et al. Preserved extracellular matrix components and retained biological activity in decellularized porcine mesothelium. Biomaterials. 2010;31:6934–40.CrossRefPubMed Hoganson DM, Owens GE, O'Doherty EM, Bowley CM, Goldman SM, Harilal DO, et al. Preserved extracellular matrix components and retained biological activity in decellularized porcine mesothelium. Biomaterials. 2010;31:6934–40.CrossRefPubMed
55.
go back to reference Sicari BM, Dziki JL, Badylak SF. Strategies for functional bioscaffold-based skeletal muscle reconstruction. Ann Transl Med. 2015;3:256.PubMedPubMedCentral Sicari BM, Dziki JL, Badylak SF. Strategies for functional bioscaffold-based skeletal muscle reconstruction. Ann Transl Med. 2015;3:256.PubMedPubMedCentral
56.
go back to reference Turner NJ, Badylak SF. Biologic scaffolds for musculotendinous tissue repair. Eur Cell Mater. 2013;25:130–43.PubMed Turner NJ, Badylak SF. Biologic scaffolds for musculotendinous tissue repair. Eur Cell Mater. 2013;25:130–43.PubMed
57.
go back to reference Agrawal V, Brown BN, Beattie AJ, Gilbert TW, Badylak SF. Evidence of innervation following extracellular matrix scaffold-mediated remodelling of muscular tissues. J Tissue Eng Regen Med. 2009;3:590–600. John Wiley & Sons, Ltd.CrossRefPubMedPubMedCentral Agrawal V, Brown BN, Beattie AJ, Gilbert TW, Badylak SF. Evidence of innervation following extracellular matrix scaffold-mediated remodelling of muscular tissues. J Tissue Eng Regen Med. 2009;3:590–600. John Wiley & Sons, Ltd.CrossRefPubMedPubMedCentral
58.
go back to reference Sicari BM, Agrawal V, Siu BF, Medberry CJ, Dearth CL, Turner NJ, et al. A murine model of volumetric muscle loss and a regenerative medicine approach for tissue replacement. Tissue Eng Part A. 2012;18:1941–8.CrossRefPubMedPubMedCentral Sicari BM, Agrawal V, Siu BF, Medberry CJ, Dearth CL, Turner NJ, et al. A murine model of volumetric muscle loss and a regenerative medicine approach for tissue replacement. Tissue Eng Part A. 2012;18:1941–8.CrossRefPubMedPubMedCentral
59.
go back to reference Turner NJ, Badylak JS, Weber DJ, Badylak SF. Biologic scaffold remodeling in a dog model of complex musculoskeletal injury. J Surg Res. 2012;176:490–502. Elsevier.CrossRefPubMed Turner NJ, Badylak JS, Weber DJ, Badylak SF. Biologic scaffold remodeling in a dog model of complex musculoskeletal injury. J Surg Res. 2012;176:490–502. Elsevier.CrossRefPubMed
60.
go back to reference Murakami K, Aoki H, Nakamura S, Nakamura S-I, Takikawa M, Hanzawa M, et al. Hydrogel blends of chitin/chitosan, fucoidan and alginate as healing-impaired wound dressings. Biomaterials. 2010;31:83–90.CrossRefPubMed Murakami K, Aoki H, Nakamura S, Nakamura S-I, Takikawa M, Hanzawa M, et al. Hydrogel blends of chitin/chitosan, fucoidan and alginate as healing-impaired wound dressings. Biomaterials. 2010;31:83–90.CrossRefPubMed
62.
go back to reference Khaing ZZ, Schmidt CE. Advances in natural biomaterials for nerve tissue repair. Neurosci Lett. 2012;519:103–14.CrossRefPubMed Khaing ZZ, Schmidt CE. Advances in natural biomaterials for nerve tissue repair. Neurosci Lett. 2012;519:103–14.CrossRefPubMed
63.
go back to reference Bidarra SJ, Barrias CC, Granja PL. Injectable alginate hydrogels for cell delivery in tissue engineering. Acta Biomater. 2014;10:1646–62.CrossRefPubMed Bidarra SJ, Barrias CC, Granja PL. Injectable alginate hydrogels for cell delivery in tissue engineering. Acta Biomater. 2014;10:1646–62.CrossRefPubMed
64.
go back to reference Walters BD, Stegemann JP. Strategies for directing the structure and function of three-dimensional collagen biomaterials across length scales. Acta Biomater. 2014;10:1488–501.CrossRefPubMed Walters BD, Stegemann JP. Strategies for directing the structure and function of three-dimensional collagen biomaterials across length scales. Acta Biomater. 2014;10:1488–501.CrossRefPubMed
66.
go back to reference Brown AC, Barker TH. Fibrin-based biomaterials: modulation of macroscopic properties through rational design at the molecular level. Acta Biomater. 2014;10:1502–14.CrossRefPubMed Brown AC, Barker TH. Fibrin-based biomaterials: modulation of macroscopic properties through rational design at the molecular level. Acta Biomater. 2014;10:1502–14.CrossRefPubMed
67.
go back to reference Boontheekul T, Hill EE, Kong H-J, Mooney DJ. Regulating myoblast phenotype through controlled gel stiffness and degradation. Tissue Eng. 2007;13:1431–42.CrossRefPubMed Boontheekul T, Hill EE, Kong H-J, Mooney DJ. Regulating myoblast phenotype through controlled gel stiffness and degradation. Tissue Eng. 2007;13:1431–42.CrossRefPubMed
68.
go back to reference Hill E, Boontheekul T, Mooney DJ. Designing scaffolds to enhance transplanted myoblast survival and migration. Tissue Eng. 2006;12:1295–304.CrossRefPubMed Hill E, Boontheekul T, Mooney DJ. Designing scaffolds to enhance transplanted myoblast survival and migration. Tissue Eng. 2006;12:1295–304.CrossRefPubMed
69.
go back to reference Hill E, Boontheekul T, Mooney DJ. Regulating activation of transplanted cells controls tissue regeneration. Proc Natl Acad Sci U S A. 2006;103:2494–9. National Acad Sciences.CrossRefPubMedPubMedCentral Hill E, Boontheekul T, Mooney DJ. Regulating activation of transplanted cells controls tissue regeneration. Proc Natl Acad Sci U S A. 2006;103:2494–9. National Acad Sciences.CrossRefPubMedPubMedCentral
70.
go back to reference Borselli C, Storrie H, Benesch-Lee F, Shvartsman D, Cezar C, Lichtman JW, et al. Functional muscle regeneration with combined delivery of angiogenesis and myogenesis factors. Proc Natl Acad Sci U S A. 2010;107:3287–92. National Acad Sciences.CrossRefPubMedPubMedCentral Borselli C, Storrie H, Benesch-Lee F, Shvartsman D, Cezar C, Lichtman JW, et al. Functional muscle regeneration with combined delivery of angiogenesis and myogenesis factors. Proc Natl Acad Sci U S A. 2010;107:3287–92. National Acad Sciences.CrossRefPubMedPubMedCentral
71.
go back to reference Shandalov Y, Egozi D, Koffler J, Dado-Rosenfeld D, Ben-Shimol D, Freiman A, et al. An engineered muscle flap for reconstruction of large soft tissue defects. Proc Natl Acad Sci U S A. 2014;111:6010–5. National Acad Sciences.CrossRefPubMedPubMedCentral Shandalov Y, Egozi D, Koffler J, Dado-Rosenfeld D, Ben-Shimol D, Freiman A, et al. An engineered muscle flap for reconstruction of large soft tissue defects. Proc Natl Acad Sci U S A. 2014;111:6010–5. National Acad Sciences.CrossRefPubMedPubMedCentral
72.
go back to reference Boldrin L, Elvassore N, Malerba A, Flaibani M, Cimetta E, Piccoli M, et al. Satellite cells delivered by micro-patterned scaffolds: a new strategy for cell transplantation in muscle diseases. Tissue Eng. 2007;13:253–62.CrossRefPubMed Boldrin L, Elvassore N, Malerba A, Flaibani M, Cimetta E, Piccoli M, et al. Satellite cells delivered by micro-patterned scaffolds: a new strategy for cell transplantation in muscle diseases. Tissue Eng. 2007;13:253–62.CrossRefPubMed
73.
go back to reference Yang HS, Ieronimakis N, Tsui JH, Kim HN, Suh K-Y, Reyes M, et al. Nanopatterned muscle cell patches for enhanced myogenesis and dystrophin expression in a mouse model of muscular dystrophy. Biomaterials. 2014;35:1478–86.CrossRefPubMed Yang HS, Ieronimakis N, Tsui JH, Kim HN, Suh K-Y, Reyes M, et al. Nanopatterned muscle cell patches for enhanced myogenesis and dystrophin expression in a mouse model of muscular dystrophy. Biomaterials. 2014;35:1478–86.CrossRefPubMed
74.
go back to reference Almany L, Seliktar D. Biosynthetic hydrogel scaffolds made from fibrinogen and polyethylene glycol for 3D cell cultures. Biomaterials. 2005;26:2467–77.CrossRefPubMed Almany L, Seliktar D. Biosynthetic hydrogel scaffolds made from fibrinogen and polyethylene glycol for 3D cell cultures. Biomaterials. 2005;26:2467–77.CrossRefPubMed
75.
go back to reference Peyton SR, Kim PD, Ghajar CM, Seliktar D, Putnam AJ. The effects of matrix stiffness and RhoA on the phenotypic plasticity of smooth muscle cells in a 3-D biosynthetic hydrogel system. Biomaterials. 2008;29:2597–607.CrossRefPubMedPubMedCentral Peyton SR, Kim PD, Ghajar CM, Seliktar D, Putnam AJ. The effects of matrix stiffness and RhoA on the phenotypic plasticity of smooth muscle cells in a 3-D biosynthetic hydrogel system. Biomaterials. 2008;29:2597–607.CrossRefPubMedPubMedCentral
76.
go back to reference Fuoco C, Salvatori ML, Biondo A, Shapira-Schweitzer K, Santoleri S, Antonini S, et al. Injectable polyethylene glycol-fibrinogen hydrogel adjuvant improves survival and differentiation of transplanted mesoangioblasts in acute and chronic skeletal-muscle degeneration. Skelet Muscle. 2012;2:24. BioMed Central.CrossRefPubMedPubMedCentral Fuoco C, Salvatori ML, Biondo A, Shapira-Schweitzer K, Santoleri S, Antonini S, et al. Injectable polyethylene glycol-fibrinogen hydrogel adjuvant improves survival and differentiation of transplanted mesoangioblasts in acute and chronic skeletal-muscle degeneration. Skelet Muscle. 2012;2:24. BioMed Central.CrossRefPubMedPubMedCentral
77.
go back to reference Fuoco C, Rizzi R, Biondo A, Longa E, Mascaro A, Shapira-Schweitzer K, et al. In vivo generation of a mature and functional artificial skeletal muscle. EMBO Mol Med. 2015;7:411–22. EMBO Press.CrossRefPubMedPubMedCentral Fuoco C, Rizzi R, Biondo A, Longa E, Mascaro A, Shapira-Schweitzer K, et al. In vivo generation of a mature and functional artificial skeletal muscle. EMBO Mol Med. 2015;7:411–22. EMBO Press.CrossRefPubMedPubMedCentral
Metadata
Title
Matrix scaffolding for stem cell guidance toward skeletal muscle tissue engineering
Authors
Claudia Fuoco
Lucia Lisa Petrilli
Stefano Cannata
Cesare Gargioli
Publication date
01-12-2016
Publisher
BioMed Central
Published in
Journal of Orthopaedic Surgery and Research / Issue 1/2016
Electronic ISSN: 1749-799X
DOI
https://doi.org/10.1186/s13018-016-0421-y

Other articles of this Issue 1/2016

Journal of Orthopaedic Surgery and Research 1/2016 Go to the issue