Skip to main content
Top
Published in: Journal of Artificial Organs 3/2012

01-09-2012 | Review

Bioactive polymer scaffold for fabrication of vascularized engineering tissue

Author: Irza Sukmana

Published in: Journal of Artificial Organs | Issue 3/2012

Login to get access

Abstract

Tissue engineering seeks strategies to design polymeric scaffolds that allow high-cell-density cultures with signaling molecules and suitable vascular supply. One major obstacle in tissue engineering is the inability to create thick engineered-tissue constructs. A pre-vascularized tissue scaffold appears to be the most favorable approach to avoid nutrient and oxygen supply limitations as well as to allow waste removal, factors that are often hurdles in developing thick engineered tissues. Vascularization can be achieved using strategies in which cells are cultured in bioactive polymer scaffolds that can mimic extracellular matrix environments. This review addresses recent advances and future challenges in developing and using bioactive polymer scaffolds to promote tissue construct vascularization.
Literature
1.
go back to reference Atala A. Tissue engineering and regenerative medicine: concepts for clinical application. Rejuvenation Res. 2004;7:15–31.PubMedCrossRef Atala A. Tissue engineering and regenerative medicine: concepts for clinical application. Rejuvenation Res. 2004;7:15–31.PubMedCrossRef
2.
go back to reference Orlando G, Wood KJ, Stratta RJ, Atala A, Soker S. Regenerative medicine and organ transplantation: past, present, and future. Transplantation. 2011;91:1310–7.PubMedCrossRef Orlando G, Wood KJ, Stratta RJ, Atala A, Soker S. Regenerative medicine and organ transplantation: past, present, and future. Transplantation. 2011;91:1310–7.PubMedCrossRef
4.
go back to reference Neumann T, Nicholson BS, Sanders JE. Tissue engineering of perfused microvessels. Microvasc Res. 2003;66:59–67.PubMedCrossRef Neumann T, Nicholson BS, Sanders JE. Tissue engineering of perfused microvessels. Microvasc Res. 2003;66:59–67.PubMedCrossRef
5.
6.
go back to reference Mikos AG, Sarakinos G, Lyman MD, Ingber DE, Vacanti JP, Langer R. Prevascularization of porous biodegradable polymers. Biotechnol Bioeng. 1993;42:716–23.PubMedCrossRef Mikos AG, Sarakinos G, Lyman MD, Ingber DE, Vacanti JP, Langer R. Prevascularization of porous biodegradable polymers. Biotechnol Bioeng. 1993;42:716–23.PubMedCrossRef
7.
8.
go back to reference Nerem RM. Cell-based therapies: from basic biology to replacement, repair, and regeneration. Biomaterials. 2007;28:5074–7.PubMedCrossRef Nerem RM. Cell-based therapies: from basic biology to replacement, repair, and regeneration. Biomaterials. 2007;28:5074–7.PubMedCrossRef
9.
go back to reference He W, Ye L, Li S, Liu H, Wu B, Wang Q, et al. Construction of vascularized cardiac tissue from genetically modified mouse embryonic stem cells. J Heart Lung Transplant. 2012;31:204–12.PubMedCrossRef He W, Ye L, Li S, Liu H, Wu B, Wang Q, et al. Construction of vascularized cardiac tissue from genetically modified mouse embryonic stem cells. J Heart Lung Transplant. 2012;31:204–12.PubMedCrossRef
10.
go back to reference Koh CJ, Delo DM, Lee JW, Siddiqui MM, Lanza RP, Soker S, Yoo JJ, Atala A. Parthenogenesis-derived multipotent stem cells adapted for tissue engineering applications. Methods. 2009;47:90–7.PubMedCrossRef Koh CJ, Delo DM, Lee JW, Siddiqui MM, Lanza RP, Soker S, Yoo JJ, Atala A. Parthenogenesis-derived multipotent stem cells adapted for tissue engineering applications. Methods. 2009;47:90–7.PubMedCrossRef
11.
go back to reference Conway EM, Carmeliet P. The diversity of endothelial cells: a challenge for therapeutic angiogenesis. Genome Biol. 2004;5:207.PubMedCrossRef Conway EM, Carmeliet P. The diversity of endothelial cells: a challenge for therapeutic angiogenesis. Genome Biol. 2004;5:207.PubMedCrossRef
12.
go back to reference Gimble J, Guilak F. Adipose-derived adult stem cells: isolation, characterization, and differentiation potential. Cytotherapy. 2003;5:362–9.PubMedCrossRef Gimble J, Guilak F. Adipose-derived adult stem cells: isolation, characterization, and differentiation potential. Cytotherapy. 2003;5:362–9.PubMedCrossRef
13.
14.
go back to reference Howe A, Aplin AE, Alahari SK, Juliano RL. Integrin signaling and cell growth control. Curr Opin Cell Biol. 1998;10:220–3.PubMedCrossRef Howe A, Aplin AE, Alahari SK, Juliano RL. Integrin signaling and cell growth control. Curr Opin Cell Biol. 1998;10:220–3.PubMedCrossRef
15.
go back to reference Frescaline G, Bouderlique T, Huynh MB, Papy-Garcia D, Courty J, Albanese P. Glycosaminoglycans mimetics potentiate the clonogenicity, proliferation, migration and differentiation properties of rat mesenchymal stem cells. Stem Cell Res. 2012;8:180–92.PubMedCrossRef Frescaline G, Bouderlique T, Huynh MB, Papy-Garcia D, Courty J, Albanese P. Glycosaminoglycans mimetics potentiate the clonogenicity, proliferation, migration and differentiation properties of rat mesenchymal stem cells. Stem Cell Res. 2012;8:180–92.PubMedCrossRef
16.
go back to reference Chang C, Werb Z. The many faces of metalloproteases: cell growth, invasion, angiogenesis and metastasis. Trends Cell Biol. 2001;11:S37–43.PubMed Chang C, Werb Z. The many faces of metalloproteases: cell growth, invasion, angiogenesis and metastasis. Trends Cell Biol. 2001;11:S37–43.PubMed
18.
go back to reference Lee CH, Huang GS, Chao KH, Wu SS, Chen Q. Differential pretensions of a flexor tendon graft for anterior cruciate ligament reconstruction: a biomechanical comparison in a porcine knee model. Arthroscopy. 2005;21:540–6.PubMedCrossRef Lee CH, Huang GS, Chao KH, Wu SS, Chen Q. Differential pretensions of a flexor tendon graft for anterior cruciate ligament reconstruction: a biomechanical comparison in a porcine knee model. Arthroscopy. 2005;21:540–6.PubMedCrossRef
19.
go back to reference Fuchs JR, Nasseri BA, Vacanti JP. Tissue engineering: a 21st century solution to surgical reconstruction. Ann Thorac Surg. 2001;72:577–91.PubMedCrossRef Fuchs JR, Nasseri BA, Vacanti JP. Tissue engineering: a 21st century solution to surgical reconstruction. Ann Thorac Surg. 2001;72:577–91.PubMedCrossRef
20.
go back to reference Chaikof EL, Matthew H, Kohn J, Mikos AG, Prestwich GD, Yip CM. Biomaterials and scaffolds in reparative medicine. Ann N Y Acad Sci. 2002;961:96–105.PubMedCrossRef Chaikof EL, Matthew H, Kohn J, Mikos AG, Prestwich GD, Yip CM. Biomaterials and scaffolds in reparative medicine. Ann N Y Acad Sci. 2002;961:96–105.PubMedCrossRef
21.
go back to reference Larson JW III, Chu CR. Tissue engineering of articular cartilage. In: Guelcher SA, Hollinger JO, editors. An introduction to biomaterials. 2nd ed. Boca Raton: CRC Press; 2006. p. 525–36. Larson JW III, Chu CR. Tissue engineering of articular cartilage. In: Guelcher SA, Hollinger JO, editors. An introduction to biomaterials. 2nd ed. Boca Raton: CRC Press; 2006. p. 525–36.
22.
go back to reference Mooney DJ, Sano K, Kaufmann PM, Majahod K, Schloo B, Vacanti JP, Langer R. Long-term engraftment of hepatocytes transplanted on biodegradable polymer sponges. J Biomed Mater Res. 1997;37:413–20.PubMedCrossRef Mooney DJ, Sano K, Kaufmann PM, Majahod K, Schloo B, Vacanti JP, Langer R. Long-term engraftment of hepatocytes transplanted on biodegradable polymer sponges. J Biomed Mater Res. 1997;37:413–20.PubMedCrossRef
23.
go back to reference Tabata Y. Biomaterial technology for tissue engineering applications. J R Soc Interface. 2009;6:S311–24.PubMedCrossRef Tabata Y. Biomaterial technology for tissue engineering applications. J R Soc Interface. 2009;6:S311–24.PubMedCrossRef
24.
go back to reference Lee JW, Lan PX, Kim B, Lim G, Cho DW. Fabrication and characteristic analysis of a poly(propylene fumarate) scaffold using micro-stereolithography technology. J Biomed Mater Res B Appl Biomater. 2008;87:1–9.PubMed Lee JW, Lan PX, Kim B, Lim G, Cho DW. Fabrication and characteristic analysis of a poly(propylene fumarate) scaffold using micro-stereolithography technology. J Biomed Mater Res B Appl Biomater. 2008;87:1–9.PubMed
25.
go back to reference Kikuchi A, Okamoto S, Takahashi S, Asano S, Nishikawa T. Linear chronic cutaneous graft-versus-host disease. J Am Acad Dermatol. 1997;37:1004–6.PubMedCrossRef Kikuchi A, Okamoto S, Takahashi S, Asano S, Nishikawa T. Linear chronic cutaneous graft-versus-host disease. J Am Acad Dermatol. 1997;37:1004–6.PubMedCrossRef
26.
go back to reference Vert M, Mauduit J, Li S. Biodegradation of PLA/GA polymers: increasing complexity. Biomaterials. 1994;15:1209–13.PubMedCrossRef Vert M, Mauduit J, Li S. Biodegradation of PLA/GA polymers: increasing complexity. Biomaterials. 1994;15:1209–13.PubMedCrossRef
27.
go back to reference Bramfeldt H, Sarazin P, Vermette P. Characterization, degradation, and mechanical strength of poly(d,l-lactide-co-epsilon-caprolactone)-poly(ethylene glycol)-poly(d,l-lactide-co-epsilon-caprolactone). J Biomed Mater Res A. 2007;83:503–11.PubMed Bramfeldt H, Sarazin P, Vermette P. Characterization, degradation, and mechanical strength of poly(d,l-lactide-co-epsilon-caprolactone)-poly(ethylene glycol)-poly(d,l-lactide-co-epsilon-caprolactone). J Biomed Mater Res A. 2007;83:503–11.PubMed
28.
go back to reference Grizzi I, Garreau H, Li S, Vert M. Hydrolytic degradation of devices based on poly(dl-lactic acid) size-dependence. Biomaterials. 1995;16:305–11.PubMedCrossRef Grizzi I, Garreau H, Li S, Vert M. Hydrolytic degradation of devices based on poly(dl-lactic acid) size-dependence. Biomaterials. 1995;16:305–11.PubMedCrossRef
29.
go back to reference Levenberg S, Rouwkema J, Macdonald M, Garfein ES, Kohane DS, Darland DC, Marini R, van Blitterswijk CA, Mulligan RC, D’Amore PA, Langer R. Engineering vascularized skeletal muscle tissue. Nat Biotechnol. 2005;23:879–84.PubMedCrossRef Levenberg S, Rouwkema J, Macdonald M, Garfein ES, Kohane DS, Darland DC, Marini R, van Blitterswijk CA, Mulligan RC, D’Amore PA, Langer R. Engineering vascularized skeletal muscle tissue. Nat Biotechnol. 2005;23:879–84.PubMedCrossRef
30.
go back to reference Pego AP, Siebum B, Van Luyn MJ. Preparation of degradable porous structures based on 1,3-trimethylene carbonate and d,l-lactide (co)polymers for heart tissue engineering. Tissue Eng. 2003;9:981–94.PubMedCrossRef Pego AP, Siebum B, Van Luyn MJ. Preparation of degradable porous structures based on 1,3-trimethylene carbonate and d,l-lactide (co)polymers for heart tissue engineering. Tissue Eng. 2003;9:981–94.PubMedCrossRef
31.
go back to reference Pego AP, Van Luyn MJ, Brouwer LA, van Wachem PB, Poot AA, Grijpma DW, Feijen J. In vivo behavior of poly(1,3-trimethylene carbonate) and copolymers of 1,3-trimethylene carbonate with d,l-lactide or epsilon-caprolactone: degradation and tissue response. J Biomed Mater Res A. 2003;67:1044–54.PubMedCrossRef Pego AP, Van Luyn MJ, Brouwer LA, van Wachem PB, Poot AA, Grijpma DW, Feijen J. In vivo behavior of poly(1,3-trimethylene carbonate) and copolymers of 1,3-trimethylene carbonate with d,l-lactide or epsilon-caprolactone: degradation and tissue response. J Biomed Mater Res A. 2003;67:1044–54.PubMedCrossRef
32.
go back to reference Aird WC. Phenotypic heterogeneity of the endothelium: II. Representative vascular beds. Circ Res. 2007;100:158–73.PubMedCrossRef Aird WC. Phenotypic heterogeneity of the endothelium: II. Representative vascular beds. Circ Res. 2007;100:158–73.PubMedCrossRef
33.
go back to reference Hutchings H, Ortega N, Plouet J. Extracellular matrix-bound vascular endothelial growth factor promotes endothelial cell adhesion, migration, and survival through integrin ligation. FASEB J. 2003;17:1520–2.PubMed Hutchings H, Ortega N, Plouet J. Extracellular matrix-bound vascular endothelial growth factor promotes endothelial cell adhesion, migration, and survival through integrin ligation. FASEB J. 2003;17:1520–2.PubMed
34.
go back to reference Brekke JH, Goldman SM, Ieska K, Issack P, Bong MR, Tian H. Hyaluronan as a biomaterials. In: Guelcher SA, Hollinger JO, editors. An introduction to biomaterials. 2nd ed. Boca Raton: CRC Press; 2006. p. 219–48. Brekke JH, Goldman SM, Ieska K, Issack P, Bong MR, Tian H. Hyaluronan as a biomaterials. In: Guelcher SA, Hollinger JO, editors. An introduction to biomaterials. 2nd ed. Boca Raton: CRC Press; 2006. p. 219–48.
35.
go back to reference Toole BP. Hyaluronan: from extracellular glue to pericellular cue. Nat Rev Cancer. 2004;4:528–39.PubMedCrossRef Toole BP. Hyaluronan: from extracellular glue to pericellular cue. Nat Rev Cancer. 2004;4:528–39.PubMedCrossRef
36.
go back to reference Rao CM, Deb TB, Gupta S, Datta K. Regulation of cellular phosphorylation of hyaluronan binding protein and its role in the formation of second messenger. Biochim Biophys Acta. 1997;1336:387–93.PubMedCrossRef Rao CM, Deb TB, Gupta S, Datta K. Regulation of cellular phosphorylation of hyaluronan binding protein and its role in the formation of second messenger. Biochim Biophys Acta. 1997;1336:387–93.PubMedCrossRef
37.
go back to reference Lesley J, Hascall VC, Tammi M, Hyman R. Hyaluronan binding by cell surface CD44. J Biol Chem. 2000;275:26967–75.PubMed Lesley J, Hascall VC, Tammi M, Hyman R. Hyaluronan binding by cell surface CD44. J Biol Chem. 2000;275:26967–75.PubMed
38.
go back to reference Slevin M, Kumar S, Gaffney J. Angiogenic oligosaccharides of hyaluronan induce multiple signaling pathways affecting vascular endothelial cell mitogenic and wound healing responses. J Biol Chem. 2002;277:41046–59.PubMedCrossRef Slevin M, Kumar S, Gaffney J. Angiogenic oligosaccharides of hyaluronan induce multiple signaling pathways affecting vascular endothelial cell mitogenic and wound healing responses. J Biol Chem. 2002;277:41046–59.PubMedCrossRef
39.
go back to reference Miletti-González KE, Chen S, Muthukumaran N, Saglimbeni GN, Wu X, Yang J, Apolito K, Shih WJ, Hait WN, Rodríguez-Rodríguez L. The CD44 receptor interacts with P-glycoprotein to promote cell migration and invasion in cancer. Cancer Res. 2005;65:6660–7.PubMedCrossRef Miletti-González KE, Chen S, Muthukumaran N, Saglimbeni GN, Wu X, Yang J, Apolito K, Shih WJ, Hait WN, Rodríguez-Rodríguez L. The CD44 receptor interacts with P-glycoprotein to promote cell migration and invasion in cancer. Cancer Res. 2005;65:6660–7.PubMedCrossRef
40.
go back to reference Rauh F, Dornish M. Chitosan. In: Guelcher SA, Hollinger JO, editors. An introduction to biomaterials. 2nd ed. Boca Raton: CRC Press; 2006. p. 249–59. Rauh F, Dornish M. Chitosan. In: Guelcher SA, Hollinger JO, editors. An introduction to biomaterials. 2nd ed. Boca Raton: CRC Press; 2006. p. 249–59.
41.
go back to reference Park YD, Tirelli N, Hubbell JA. Photopolymerized hyaluronic acid-based hydrogels and interpenetrating networks. Biomaterials. 2003;24:893–900.PubMedCrossRef Park YD, Tirelli N, Hubbell JA. Photopolymerized hyaluronic acid-based hydrogels and interpenetrating networks. Biomaterials. 2003;24:893–900.PubMedCrossRef
42.
go back to reference Zielinski BA, Aebischer P. Chitosan as a matrix for mammalian cell encapsulation. Biomaterials. 1994;15:1049–56.PubMedCrossRef Zielinski BA, Aebischer P. Chitosan as a matrix for mammalian cell encapsulation. Biomaterials. 1994;15:1049–56.PubMedCrossRef
43.
go back to reference Black AF, Hudon V, Damour O, Germain L, Auger FA. A novel approach for studying angiogenesis: a human skin equivalent with a capillary-like network. Cell Biol Toxicol. 1999;15:81–90.PubMedCrossRef Black AF, Hudon V, Damour O, Germain L, Auger FA. A novel approach for studying angiogenesis: a human skin equivalent with a capillary-like network. Cell Biol Toxicol. 1999;15:81–90.PubMedCrossRef
44.
go back to reference Draget KI, Skjak-Braek G, Smidsrod O. Alginate based new materials. Int J Biol Macromol. 1997;21:47–55.PubMedCrossRef Draget KI, Skjak-Braek G, Smidsrod O. Alginate based new materials. Int J Biol Macromol. 1997;21:47–55.PubMedCrossRef
45.
go back to reference Drury JL, Mooney DJ. Hydrogels for tissue engineering: scaffold design variables and applications. Biomaterials. 2003;24:4337–51.PubMedCrossRef Drury JL, Mooney DJ. Hydrogels for tissue engineering: scaffold design variables and applications. Biomaterials. 2003;24:4337–51.PubMedCrossRef
46.
go back to reference Smidsrod O, Skjak-Braek G. Alginate as immobilization matrix for cells. Trends Biotechnol. 1990;8:71–8.PubMedCrossRef Smidsrod O, Skjak-Braek G. Alginate as immobilization matrix for cells. Trends Biotechnol. 1990;8:71–8.PubMedCrossRef
47.
go back to reference Becker WM, Kleinsmith LJ, Hardin J. The world of the cell. 7th ed. San Francisco: Pearson/Benjamin Cummings; 2009. p. 229–328. Becker WM, Kleinsmith LJ, Hardin J. The world of the cell. 7th ed. San Francisco: Pearson/Benjamin Cummings; 2009. p. 229–328.
48.
go back to reference Haarer JC, Dee KC. Proteins and amino acid-derived polymers. In: Guelcher SA, Hollinger JO, editors. An introduction to biomaterials. 2nd ed. Boca Raton: CRC Press; 2006. p. 121–38. Haarer JC, Dee KC. Proteins and amino acid-derived polymers. In: Guelcher SA, Hollinger JO, editors. An introduction to biomaterials. 2nd ed. Boca Raton: CRC Press; 2006. p. 121–38.
49.
go back to reference Chen CS, Yannas IV, Spector M. Pore strain behaviour of collagen–glycosaminoglycan analogues of extracellular matrix. Biomaterials. 1995;16:777–83.PubMedCrossRef Chen CS, Yannas IV, Spector M. Pore strain behaviour of collagen–glycosaminoglycan analogues of extracellular matrix. Biomaterials. 1995;16:777–83.PubMedCrossRef
50.
go back to reference Ma L, Gao C, Mao Z, Zhou J, Shen J. Biodegradability and cell-mediated contraction of porous collagen scaffolds: the effect of lysine as a novel crosslinking bridge. J Biomed Mater Res A. 2004;71:334–42.PubMedCrossRef Ma L, Gao C, Mao Z, Zhou J, Shen J. Biodegradability and cell-mediated contraction of porous collagen scaffolds: the effect of lysine as a novel crosslinking bridge. J Biomed Mater Res A. 2004;71:334–42.PubMedCrossRef
51.
go back to reference Toman PD, Pieper F, Sakai N, Karatzas C, Platenburg E, de Wit I, Samuel C, Dekker A, Daniels GA, Berg RA, Platenburg GJ. Production of recombinant human type I procollagen homotrimer in the mammary gland of transgenic mice. Transgenic Res. 1999;8:415–27.PubMedCrossRef Toman PD, Pieper F, Sakai N, Karatzas C, Platenburg E, de Wit I, Samuel C, Dekker A, Daniels GA, Berg RA, Platenburg GJ. Production of recombinant human type I procollagen homotrimer in the mammary gland of transgenic mice. Transgenic Res. 1999;8:415–27.PubMedCrossRef
52.
go back to reference Sugiura H, Yunoki S, Kondo E, Ikoma T, Tanaka J, Yasuda K. In vivo biological responses and bioresorption of tilapia scale collagen as a potential biomaterial. J Biomater Sci Polym Ed. 2009;20:1353–68.PubMedCrossRef Sugiura H, Yunoki S, Kondo E, Ikoma T, Tanaka J, Yasuda K. In vivo biological responses and bioresorption of tilapia scale collagen as a potential biomaterial. J Biomater Sci Polym Ed. 2009;20:1353–68.PubMedCrossRef
53.
go back to reference Soker S, Machado M, Atala A. Systems for therapeutic angiogenesis in tissue engineering. World J Urol. 2000;18:10–8.PubMedCrossRef Soker S, Machado M, Atala A. Systems for therapeutic angiogenesis in tissue engineering. World J Urol. 2000;18:10–8.PubMedCrossRef
54.
go back to reference Francis ME, Uriel S, Brey EM. Endothelial cell–matrix interactions in neovascularization. Tissue Eng Part B Rev. 2008;14:19–32.PubMedCrossRef Francis ME, Uriel S, Brey EM. Endothelial cell–matrix interactions in neovascularization. Tissue Eng Part B Rev. 2008;14:19–32.PubMedCrossRef
55.
go back to reference Ingber DE, Folkman J. Mechanochemical switching between growth and differentiation during fibroblast growth factor-stimulated angiogenesis in vitro: role of extracellular matrix. J Cell Biol. 1989;109:317–30.PubMedCrossRef Ingber DE, Folkman J. Mechanochemical switching between growth and differentiation during fibroblast growth factor-stimulated angiogenesis in vitro: role of extracellular matrix. J Cell Biol. 1989;109:317–30.PubMedCrossRef
56.
go back to reference Xu J, Rodriguez D, Petitclerc E, Kim JJ, Hangai M, Moon YS, Davis GE, Brooks PC. Proteolytic exposure of a cryptic site within collagen type IV is required for angiogenesis and tumor growth in vivo. J Cell Biol. 2001;154:1069–79.PubMedCrossRef Xu J, Rodriguez D, Petitclerc E, Kim JJ, Hangai M, Moon YS, Davis GE, Brooks PC. Proteolytic exposure of a cryptic site within collagen type IV is required for angiogenesis and tumor growth in vivo. J Cell Biol. 2001;154:1069–79.PubMedCrossRef
57.
go back to reference Senger DR, Claffey KP, Benes JE, Perruzzi CA, Sergiou AP, Detmar M. Angiogenesis promoted by vascular endothelial growth factor: regulation through alpha1beta1 and alpha2beta1 integrins. Proc Natl Acad Sci USA. 1997;94:13612–7.PubMedCrossRef Senger DR, Claffey KP, Benes JE, Perruzzi CA, Sergiou AP, Detmar M. Angiogenesis promoted by vascular endothelial growth factor: regulation through alpha1beta1 and alpha2beta1 integrins. Proc Natl Acad Sci USA. 1997;94:13612–7.PubMedCrossRef
58.
go back to reference Litvinov RI, Gorkun OV, Owen SF, Shuman H, Weisel JW. Polymerization of fibrin: specificity, strength, and stability of knob-hole interactions studied at the single-molecule level. Blood. 2005;106:2944–51.PubMedCrossRef Litvinov RI, Gorkun OV, Owen SF, Shuman H, Weisel JW. Polymerization of fibrin: specificity, strength, and stability of knob-hole interactions studied at the single-molecule level. Blood. 2005;106:2944–51.PubMedCrossRef
59.
go back to reference Janmey PA, Winer JP, Weisel JW. Fibrin gels and their clinical and bioengineering applications. Interface. 2009;6:1–10.PubMedCrossRef Janmey PA, Winer JP, Weisel JW. Fibrin gels and their clinical and bioengineering applications. Interface. 2009;6:1–10.PubMedCrossRef
61.
go back to reference Rowe SL, Lee S, Stegemann JP. Influence of thrombin concentration on the mechanical and morphological properties of cell-seeded fibrin hydrogels. Acta Biomater. 2007;3:59–67.PubMedCrossRef Rowe SL, Lee S, Stegemann JP. Influence of thrombin concentration on the mechanical and morphological properties of cell-seeded fibrin hydrogels. Acta Biomater. 2007;3:59–67.PubMedCrossRef
62.
go back to reference Ferrenq I, Tranqui L, Vailhé B, Gumery PY, Tracqui P. Modelling biological gel contraction by cells: mechanocellular formulation and cell traction force quantification. Acta Biotheor. 1997;45:267–93.PubMedCrossRef Ferrenq I, Tranqui L, Vailhé B, Gumery PY, Tracqui P. Modelling biological gel contraction by cells: mechanocellular formulation and cell traction force quantification. Acta Biotheor. 1997;45:267–93.PubMedCrossRef
63.
go back to reference Vailhé B, Lecomte M, Wiernsperger N, Tranqui L. The formation of tubular structures by endothelial cells is under the control of fibrinolysis and mechanical factors. Angiogenesis. 1998;2:331–44.PubMedCrossRef Vailhé B, Lecomte M, Wiernsperger N, Tranqui L. The formation of tubular structures by endothelial cells is under the control of fibrinolysis and mechanical factors. Angiogenesis. 1998;2:331–44.PubMedCrossRef
64.
go back to reference Nehls V, Herrmann R. The configuration of fibrin clots determines capillary morphogenesis and endothelial cell migration. Microvasc Res. 1996;51:347–64.PubMedCrossRef Nehls V, Herrmann R. The configuration of fibrin clots determines capillary morphogenesis and endothelial cell migration. Microvasc Res. 1996;51:347–64.PubMedCrossRef
65.
go back to reference Cox S, Cole M, Tawil B. Behavior of human dermal fibroblasts in three-dimensional fibrin clots: dependence on fibrinogen and thrombin concentration. Tissue Eng. 2004;10:942–54.PubMedCrossRef Cox S, Cole M, Tawil B. Behavior of human dermal fibroblasts in three-dimensional fibrin clots: dependence on fibrinogen and thrombin concentration. Tissue Eng. 2004;10:942–54.PubMedCrossRef
66.
go back to reference Catelas I, Sese N, Wu BM, Dunn JC, Helgerson S, Tawil B. Human mesenchymal stem cell proliferation and osteogenic differentiation in fibrin gels in vitro. Tissue Eng. 2006;12:2385–96.PubMedCrossRef Catelas I, Sese N, Wu BM, Dunn JC, Helgerson S, Tawil B. Human mesenchymal stem cell proliferation and osteogenic differentiation in fibrin gels in vitro. Tissue Eng. 2006;12:2385–96.PubMedCrossRef
67.
go back to reference Ferguson WD, Collins LM, Smith DW. Psychophysical threshold variability in cochlear implant subjects. Hear Res. 2003;180:101–13.PubMedCrossRef Ferguson WD, Collins LM, Smith DW. Psychophysical threshold variability in cochlear implant subjects. Hear Res. 2003;180:101–13.PubMedCrossRef
68.
go back to reference Tassiopoulos AK, Golts E, Oh DS, Labropoulos N. Current concepts in chronic venous ulceration. Eur J Vasc Endovasc Surg. 2000;20:227–32.PubMedCrossRef Tassiopoulos AK, Golts E, Oh DS, Labropoulos N. Current concepts in chronic venous ulceration. Eur J Vasc Endovasc Surg. 2000;20:227–32.PubMedCrossRef
69.
go back to reference Nakatsu MN, Hughes CCW. An optimized three-dimensional in vitro model for the analysis of angiogenesis. Methods Enzymol. 2008;443:65–82.PubMedCrossRef Nakatsu MN, Hughes CCW. An optimized three-dimensional in vitro model for the analysis of angiogenesis. Methods Enzymol. 2008;443:65–82.PubMedCrossRef
70.
go back to reference Nehls V, Drenckhahn D. A novel, microcarrier-based in vitro assay for rapid and reliable quantification of three-dimensional cell migration and angiogenesis. Microvasc Res. 1995;50:311–22.PubMedCrossRef Nehls V, Drenckhahn D. A novel, microcarrier-based in vitro assay for rapid and reliable quantification of three-dimensional cell migration and angiogenesis. Microvasc Res. 1995;50:311–22.PubMedCrossRef
71.
go back to reference Montano I, Schiestl C, Schneider J, Pontiggia L, Luginbühl J, Biedermann T, Böttcher-Haberzeth S, Braziulis E, Meuli M, Reichmann E. Formation of human capillaries in vitro: the engineering of prevascularized matrices. Tissue Eng Part A. 2009;15:1–15. Montano I, Schiestl C, Schneider J, Pontiggia L, Luginbühl J, Biedermann T, Böttcher-Haberzeth S, Braziulis E, Meuli M, Reichmann E. Formation of human capillaries in vitro: the engineering of prevascularized matrices. Tissue Eng Part A. 2009;15:1–15.
72.
73.
go back to reference Yancopoulos GD, Davis S, Gale NW, Rudge JS, Wiegand SJ, Holash J. Vascular-specific growth factors and blood vessel formation. Nature. 2000;407:242–8.PubMedCrossRef Yancopoulos GD, Davis S, Gale NW, Rudge JS, Wiegand SJ, Holash J. Vascular-specific growth factors and blood vessel formation. Nature. 2000;407:242–8.PubMedCrossRef
74.
go back to reference Chen Z, Htay A, Dos Santos W, Gillies GT, Fillmore HL, Sholley MM, Broaddus WC. In vitro angiogenesis by human umbilical vein cells (HUVEC) induced by three-dimensional co-culture with glioblastoma cells. J Neurooncol. 2009;92:121–8.PubMedCrossRef Chen Z, Htay A, Dos Santos W, Gillies GT, Fillmore HL, Sholley MM, Broaddus WC. In vitro angiogenesis by human umbilical vein cells (HUVEC) induced by three-dimensional co-culture with glioblastoma cells. J Neurooncol. 2009;92:121–8.PubMedCrossRef
75.
go back to reference Nakatsu MN, Sainson RC, Aoto JN, Taylor KL, Aitkenhead M, Pérez-del-Pulgar S, Carpenter PM, Hughes CC. Angiogenic sprouting and capillary lumen formation modeled by human umbilical vein endothelial cells (HUVEC) in fibrin gels: the role of fibroblasts and angiopoietin-1. Microvasc Res. 2003;66:102–12.PubMedCrossRef Nakatsu MN, Sainson RC, Aoto JN, Taylor KL, Aitkenhead M, Pérez-del-Pulgar S, Carpenter PM, Hughes CC. Angiogenic sprouting and capillary lumen formation modeled by human umbilical vein endothelial cells (HUVEC) in fibrin gels: the role of fibroblasts and angiopoietin-1. Microvasc Res. 2003;66:102–12.PubMedCrossRef
76.
go back to reference Sukmana I, Vermette P. Polymer fibers as contact guidance to orient microvascularization in a 3D environment. J Biomed Mater Res A. 2010;92:1587–97.PubMed Sukmana I, Vermette P. Polymer fibers as contact guidance to orient microvascularization in a 3D environment. J Biomed Mater Res A. 2010;92:1587–97.PubMed
77.
go back to reference Sukmana I, Vermette P. The effect of co-culture with fibroblast and angiogenic growth factors on microvascular maturation and multi-cellular lumen formation in HUVEC-oriented polymer fibre constructs. Biomaterials. 2010;31:5100–9.CrossRef Sukmana I, Vermette P. The effect of co-culture with fibroblast and angiogenic growth factors on microvascular maturation and multi-cellular lumen formation in HUVEC-oriented polymer fibre constructs. Biomaterials. 2010;31:5100–9.CrossRef
78.
go back to reference Lishko VK, Kudryk B, Yakubenko VP, Yee VC, Ugarova TP. Regulated unmasking of the cryptic binding site for integrin alpha M beta 2 in the gamma C-domain of fibrinogen. Biochemistry. 2002;41:12942–51.PubMedCrossRef Lishko VK, Kudryk B, Yakubenko VP, Yee VC, Ugarova TP. Regulated unmasking of the cryptic binding site for integrin alpha M beta 2 in the gamma C-domain of fibrinogen. Biochemistry. 2002;41:12942–51.PubMedCrossRef
79.
go back to reference Cheresh DA, Berliner SA, Vicente V, Ruggeri ZM. Recognition of distinct adhesive sites on fibrinogen by related integrins on platelets and endothelial cells. Cell. 1989;58:945–53.PubMedCrossRef Cheresh DA, Berliner SA, Vicente V, Ruggeri ZM. Recognition of distinct adhesive sites on fibrinogen by related integrins on platelets and endothelial cells. Cell. 1989;58:945–53.PubMedCrossRef
81.
go back to reference Monchaux E, Vermette P. Effects of surface properties and bioactivation of biomaterials on endothelial cells. Front Biosci (Schol Ed). 2010;2:239–55.CrossRef Monchaux E, Vermette P. Effects of surface properties and bioactivation of biomaterials on endothelial cells. Front Biosci (Schol Ed). 2010;2:239–55.CrossRef
82.
go back to reference Bayless KJ, Davis GE. Sphingosine-1-phosphate markedly induces matrix metalloproteinase and integrin-dependent human endothelial cell invasion and lumen formation in three-dimensional collagen and fibrin matrices. Biochem Biophys Res Commun. 2003;312:903–13.PubMedCrossRef Bayless KJ, Davis GE. Sphingosine-1-phosphate markedly induces matrix metalloproteinase and integrin-dependent human endothelial cell invasion and lumen formation in three-dimensional collagen and fibrin matrices. Biochem Biophys Res Commun. 2003;312:903–13.PubMedCrossRef
83.
go back to reference Isenberg BC, Williams C, Tranquillo RT. Small-diameter artificial arteries engineered in vitro. Circ Res. 2006;98:25–35.PubMedCrossRef Isenberg BC, Williams C, Tranquillo RT. Small-diameter artificial arteries engineered in vitro. Circ Res. 2006;98:25–35.PubMedCrossRef
84.
go back to reference Mooney DJ, Organ G, Vacanti JP, Langer R. Design and fabrication of biodegradable polymer devices to engineer tubular tissues. Cell Transplant. 1994;3:203–10.PubMed Mooney DJ, Organ G, Vacanti JP, Langer R. Design and fabrication of biodegradable polymer devices to engineer tubular tissues. Cell Transplant. 1994;3:203–10.PubMed
85.
go back to reference Mooney DJ, Mazzoni CL, Breuer C, McNamara K, Hern D, Vacanti JP, Langer R. Stabilized polyglycolic acid fibre-based tubes for tissue engineering. Biomaterials. 1996;17:115–24.PubMedCrossRef Mooney DJ, Mazzoni CL, Breuer C, McNamara K, Hern D, Vacanti JP, Langer R. Stabilized polyglycolic acid fibre-based tubes for tissue engineering. Biomaterials. 1996;17:115–24.PubMedCrossRef
86.
go back to reference Lesman A, Blinder Y, Levenberg Y. Modeling of flow-induced shear stress applied on 3D cellular scaffolds: implications for vascular tissue engineering. Biotechnol Bioeng. 2010;105:645–54.PubMedCrossRef Lesman A, Blinder Y, Levenberg Y. Modeling of flow-induced shear stress applied on 3D cellular scaffolds: implications for vascular tissue engineering. Biotechnol Bioeng. 2010;105:645–54.PubMedCrossRef
87.
go back to reference Davis GE, Black SM, Bayless KJ. Capillary morphogenesis during human endothelial cell invasion of three-dimensional collagen matrices. In Vitro Cell Dev Biol Anim. 2000;36:513–9.PubMedCrossRef Davis GE, Black SM, Bayless KJ. Capillary morphogenesis during human endothelial cell invasion of three-dimensional collagen matrices. In Vitro Cell Dev Biol Anim. 2000;36:513–9.PubMedCrossRef
88.
go back to reference Kannan RY, Salacinski HJ, Sales K, Butler P, Seifalian AM. The roles of tissue engineering and vascularisation in the development of micro-vascular networks: a review. Biomaterials. 2005;26:1857–75.PubMedCrossRef Kannan RY, Salacinski HJ, Sales K, Butler P, Seifalian AM. The roles of tissue engineering and vascularisation in the development of micro-vascular networks: a review. Biomaterials. 2005;26:1857–75.PubMedCrossRef
89.
go back to reference Agrawal CM, Ray RB. Biodegradable polymeric scaffolds for musculoskeletal tissue engineering. J Biomed Mater Res. 2001;55:141–50.PubMedCrossRef Agrawal CM, Ray RB. Biodegradable polymeric scaffolds for musculoskeletal tissue engineering. J Biomed Mater Res. 2001;55:141–50.PubMedCrossRef
90.
go back to reference Linnes MP, Ratner BD, Giachelli CM. A fibrinogen-based precision microporous scaffold for tissue engineering. Biomaterials. 2007;28:5298–306.PubMedCrossRef Linnes MP, Ratner BD, Giachelli CM. A fibrinogen-based precision microporous scaffold for tissue engineering. Biomaterials. 2007;28:5298–306.PubMedCrossRef
91.
go back to reference Clowes AW, Kirkman TR, Clowes MM. Mechanisms of arterial graft failure. II. Chronic endothelial and smooth muscle cell proliferation in healing polytetrafluoroethylene prostheses. J Vasc Surg. 1986;3:877–84.PubMed Clowes AW, Kirkman TR, Clowes MM. Mechanisms of arterial graft failure. II. Chronic endothelial and smooth muscle cell proliferation in healing polytetrafluoroethylene prostheses. J Vasc Surg. 1986;3:877–84.PubMed
92.
go back to reference O’Brien FJ, Taylor D, Clive LT. The effect of pore size on cell adhesion in collagen–GAG scaffolds. Biomaterials. 2005;26:433–41.PubMedCrossRef O’Brien FJ, Taylor D, Clive LT. The effect of pore size on cell adhesion in collagen–GAG scaffolds. Biomaterials. 2005;26:433–41.PubMedCrossRef
93.
go back to reference Zeltinger J, Sherwood JK, Graham DA, Müeller R, Griffith LG. Effect of pore size and void fraction on cellular adhesion, proliferation, and matrix deposition. Tissue Eng. 2001;7:557–72.PubMedCrossRef Zeltinger J, Sherwood JK, Graham DA, Müeller R, Griffith LG. Effect of pore size and void fraction on cellular adhesion, proliferation, and matrix deposition. Tissue Eng. 2001;7:557–72.PubMedCrossRef
94.
go back to reference Huang Y, Onyeri S, Siewe M, Moshfeghian A, Madihally SV. In vitro characterization of chitosan–gelatin scaffolds for tissue engineering. Biomaterials. 2005;26:7616–27.PubMedCrossRef Huang Y, Onyeri S, Siewe M, Moshfeghian A, Madihally SV. In vitro characterization of chitosan–gelatin scaffolds for tissue engineering. Biomaterials. 2005;26:7616–27.PubMedCrossRef
95.
go back to reference Zhang Z, Wang Z, Liu S, Kodama M. Pore size, tissue ingrowth, and endothelialization of small-diameter microporous polyurethane vascular prostheses. Biomaterials. 2004;25:177–87.PubMedCrossRef Zhang Z, Wang Z, Liu S, Kodama M. Pore size, tissue ingrowth, and endothelialization of small-diameter microporous polyurethane vascular prostheses. Biomaterials. 2004;25:177–87.PubMedCrossRef
96.
go back to reference Marshall A, Barker T, Sage E, Hauch K, Ratner B. Pore size controls angiogenesis in subcutaneously implanted porous matrices. In: 7th World Biomaterials Congress, Sydney; 2004. Marshall A, Barker T, Sage E, Hauch K, Ratner B. Pore size controls angiogenesis in subcutaneously implanted porous matrices. In: 7th World Biomaterials Congress, Sydney; 2004.
97.
go back to reference Yao L, Swartz DD, Gugino SF, Russell JA, Andreadis ST. Fibrin-based tissue-engineered blood vessels: differential effects of biomaterial and culture parameters on mechanical strength and vascular reactivity. Tissue Eng. 2005;11:991–1003.PubMedCrossRef Yao L, Swartz DD, Gugino SF, Russell JA, Andreadis ST. Fibrin-based tissue-engineered blood vessels: differential effects of biomaterial and culture parameters on mechanical strength and vascular reactivity. Tissue Eng. 2005;11:991–1003.PubMedCrossRef
98.
go back to reference Geiger B, Bershadsky A, Pankov R, Yamada KM. Transmembrane crosstalk between the extracellular matrix-cytoskeleton crosstalk. Nat Rev Mol Cell Biol. 2001;2:793–805.PubMedCrossRef Geiger B, Bershadsky A, Pankov R, Yamada KM. Transmembrane crosstalk between the extracellular matrix-cytoskeleton crosstalk. Nat Rev Mol Cell Biol. 2001;2:793–805.PubMedCrossRef
99.
go back to reference Ingber DE, Prusty D, Sun Z, Betensky H, Wang N. Cell shape, cytoskeletal mechanics and cell cycle control in angiogenesis. J Biomech. 1995;28:1471–84.PubMedCrossRef Ingber DE, Prusty D, Sun Z, Betensky H, Wang N. Cell shape, cytoskeletal mechanics and cell cycle control in angiogenesis. J Biomech. 1995;28:1471–84.PubMedCrossRef
100.
go back to reference Pelham RJ, Wang Y. Cell locomotion and focal adhesions are regulated by substrate flexibility. Proc Natl Acad Sci USA. 1997;94:13661–5.PubMedCrossRef Pelham RJ, Wang Y. Cell locomotion and focal adhesions are regulated by substrate flexibility. Proc Natl Acad Sci USA. 1997;94:13661–5.PubMedCrossRef
101.
go back to reference Tseng Q, Wang I, Duchemin-Pelletier E, Azioune A, Capri N, Gao J, et al. A new micropatterning method of soft substrates reveals that different tumorigenic signals can promote or reduce cell contraction levels. Lab Chip. 2011;11:2231–40.PubMedCrossRef Tseng Q, Wang I, Duchemin-Pelletier E, Azioune A, Capri N, Gao J, et al. A new micropatterning method of soft substrates reveals that different tumorigenic signals can promote or reduce cell contraction levels. Lab Chip. 2011;11:2231–40.PubMedCrossRef
102.
go back to reference Discher DE, Janmey P, Wang YL. Tissue cells feel and respond to the stiffness of their substrate. Science. 2005;310:1139–43.PubMedCrossRef Discher DE, Janmey P, Wang YL. Tissue cells feel and respond to the stiffness of their substrate. Science. 2005;310:1139–43.PubMedCrossRef
103.
go back to reference Lo CM, Wang HB, Dembo M, Wang YL. Cell movement is guided by the rigidity of the substrate. Biophys J. 2000;79:144–52.PubMedCrossRef Lo CM, Wang HB, Dembo M, Wang YL. Cell movement is guided by the rigidity of the substrate. Biophys J. 2000;79:144–52.PubMedCrossRef
104.
go back to reference Deroanne CF, Lapiere CM, Nusgens BV. In vitro tubulogenesis of endothelial cells by relaxation of the coupling extracellular matrix-cytoskeleton. Cardiovasc Res. 2001;49:647–58.PubMedCrossRef Deroanne CF, Lapiere CM, Nusgens BV. In vitro tubulogenesis of endothelial cells by relaxation of the coupling extracellular matrix-cytoskeleton. Cardiovasc Res. 2001;49:647–58.PubMedCrossRef
105.
go back to reference Yeung T, Georges PC, Flanagan LA, Marg B, Ortiz M, Funaki M, Zahir N, Ming W, Weaver V, Janmey PA. Effects of substrate stiffness on cell morphology, cytoskeletal structure, and adhesion. Cell Motil Cytoskelet. 2005;60:24–34.CrossRef Yeung T, Georges PC, Flanagan LA, Marg B, Ortiz M, Funaki M, Zahir N, Ming W, Weaver V, Janmey PA. Effects of substrate stiffness on cell morphology, cytoskeletal structure, and adhesion. Cell Motil Cytoskelet. 2005;60:24–34.CrossRef
106.
go back to reference Duong H, Wu B, Tawil B. Modulation of 3D fibrin matrix stiffness by intrinsic fibrinogen-thrombin compositions and by extrinsic cellular activity. Tissue Eng Part A. 2009;15:1865–76.PubMedCrossRef Duong H, Wu B, Tawil B. Modulation of 3D fibrin matrix stiffness by intrinsic fibrinogen-thrombin compositions and by extrinsic cellular activity. Tissue Eng Part A. 2009;15:1865–76.PubMedCrossRef
107.
go back to reference Man AJ, Davis EH, Itoh A, Leach JK, Bannerman P. Neurite outgrowth in fibrin gels is regulated by substrate stiffness. Tissue Eng Part A. 2011;17:2931–42.PubMedCrossRef Man AJ, Davis EH, Itoh A, Leach JK, Bannerman P. Neurite outgrowth in fibrin gels is regulated by substrate stiffness. Tissue Eng Part A. 2011;17:2931–42.PubMedCrossRef
Metadata
Title
Bioactive polymer scaffold for fabrication of vascularized engineering tissue
Author
Irza Sukmana
Publication date
01-09-2012
Publisher
Springer Japan
Published in
Journal of Artificial Organs / Issue 3/2012
Print ISSN: 1434-7229
Electronic ISSN: 1619-0904
DOI
https://doi.org/10.1007/s10047-012-0644-6

Other articles of this Issue 3/2012

Journal of Artificial Organs 3/2012 Go to the issue