Generic placeholder image

Current Gene Therapy

Editor-in-Chief

ISSN (Print): 1566-5232
ISSN (Online): 1875-5631

Review Article

Applications of Scaffolds in Tissue Engineering: Current Utilization and Future Prospective

Author(s): Shikha Yadav*, Javed Khan and Agrima Yadav

Volume 24, Issue 2, 2024

Published on: 26 October, 2023

Page: [94 - 109] Pages: 16

DOI: 10.2174/0115665232262167231012102837

Price: $65

conference banner
Abstract

Current regenerative medicine tactics focus on regenerating tissue structures pathologically modified by cell transplantation in combination with supporting scaffolds and biomolecules. Natural and synthetic polymers, bioresorbable inorganic and hybrid materials, and tissue decellularized were deemed biomaterials scaffolding because of their improved structural, mechanical, and biological abilities.Various biomaterials, existing treatment methodologies and emerging technologies in the field of Three-dimensional (3D) and hydrogel processing, and the unique fabric concerns for tissue engineering. A scaffold that acts as a transient matrix for cell proliferation and extracellular matrix deposition, with subsequent expansion, is needed to restore or regenerate the tissue. Diverse technologies are combined to produce porous tissue regenerative and tailored release of bioactive substances in applications of tissue engineering. Tissue engineering scaffolds are crucial ingredients. This paper discusses an overview of the various scaffold kinds and their material features and applications. Tabulation of the manufacturing technologies for fabric engineering and equipment, encompassing the latest fundamental and standard procedures.

Graphical Abstract

[1]
Engler AJ, Sen S, Sweeney HL, Discher DE. Matrix elasticity directs stem cell lineage specification. Cell 2006; 126(4): 677-89.
[http://dx.doi.org/10.1016/j.cell.2006.06.044] [PMID: 16923388]
[2]
Shruti S, Salinas AJ, Lusvardi G, Malavasi G, Menabue L, Vallet-Regi M. Mesoporous bioactive scaffolds prepared with cerium-, gallium- and zinc-containing glasses. Acta Biomater 2013; 9(1): 4836-44.
[http://dx.doi.org/10.1016/j.actbio.2012.09.024] [PMID: 23026489]
[3]
Nikolova MP, Chavali MS. Recent advances in biomaterials for 3D scaffolds: A review. Bioact Mater 2019; 4: 271-92.
[http://dx.doi.org/10.1016/j.bioactmat.2019.10.005] [PMID: 31709311]
[4]
Yilmaz F, Celep G, Tetik G. Nanofibers in Cosmetics. Nanofiber Research - Reaching New Heights intechopen. 2016.
[http://dx.doi.org/10.5772/64172]
[5]
Collins MN, Birkinshaw C. Hyaluronic acid based scaffolds for tissue engineering—A review. Carbohydr Polym 2013; 92(2): 1262-79.
[http://dx.doi.org/10.1016/j.carbpol.2012.10.028] [PMID: 23399155]
[6]
LeGeros RZ. Properties of osteoconductive biomaterials: Calcium phosphates. Clin Orthop Relat Res 2002; 395(395): 81-98.
[http://dx.doi.org/10.1097/00003086-200202000-00009] [PMID: 11937868]
[7]
Sachlos E, Czernuszka JT. Making tissue engineering scaffolds work. review: The application of solid freeform fabrication technology to the production of tissue engineering scaffolds. Eur Cell Mater 2003; 5: 29-40.
[http://dx.doi.org/10.22203/eCM.v005a03] [PMID: 14562270]
[8]
Griffith LG. Polymeric biomaterials. Acta Mater 2000; 48(1): 263-77.
[http://dx.doi.org/10.1016/S1359-6454(99)00299-2]
[9]
Hayashi T. Biodegradable polymers for biomedical uses. Prog Polym Sci 1994; 19(4): 663-702.
[http://dx.doi.org/10.1016/0079-6700(94)90030-2]
[10]
Nair LS, Laurencin CT. Biodegradable polymers as biomaterials. Prog Polym Sci 2007; 32(8-9): 762-98.
[http://dx.doi.org/10.1016/j.progpolymsci.2007.05.017]
[11]
Prasadh S, Wong RCW. Unraveling the mechanical strength of biomaterials used as a bone scaffold in oral and maxillofacial defects. Oral Sci Int 2018; 15(2): 48-55.
[http://dx.doi.org/10.1016/S1348-8643(18)30005-3]
[12]
Nanofibers and their applications in tissue engineering. - Abstract - Europe PMC, (n.d.). Avaialable from: https://europepmc.org/article/PMC/2426767 (accessed August 18, 2021).
[13]
Dormer NH, Singh M, Zhao L, Mohan N, Berkland CJ, Detamore MS. Osteochondral interface regeneration of the rabbit knee with macroscopic gradients of bioactive signals. J Biomed Mater Res A 2012; 100A(1): 162-70.
[http://dx.doi.org/10.1002/jbm.a.33225] [PMID: 22009693]
[14]
Laurencin CT, Attawia MA, Elgendy HE, Herbert KM. Tissue engineered bone-regeneration using degradable polymers: The formation of mineralized matrices. Bone 1996; 19(1): S93-9.
[http://dx.doi.org/10.1016/S8756-3282(96)00132-9] [PMID: 8831000]
[15]
Singh M, Sandhu B, Scurto A, Berkland C, Detamore MS. Microsphere-based scaffolds for cartilage tissue engineering: Using subcritical CO2 as a sintering agent. Acta Biomater 2010; 6(1): 137-43.
[http://dx.doi.org/10.1016/j.actbio.2009.07.042] [PMID: 19660579]
[16]
Stephens D, Li L, Robinson D, et al. Investigation of the in vitro release of gentamicin from a polyanhydride matrix. J Control Release 2000; 63(3): 305-17.
[http://dx.doi.org/10.1016/S0168-3659(99)00205-9] [PMID: 10601726]
[17]
Turnbull G, Clarke J, Picard F, et al. 3D bioactive composite scaffolds for bone tissue engineering. Bioact Mater 2018; 3(3): 278-314.
[http://dx.doi.org/10.1016/j.bioactmat.2017.10.001] [PMID: 29744467]
[18]
Gorth D, Webster TJ. Matrices for tissue engineering and regenerative medicine. In: Biomaterials for Artificial Organs 2011; pp. 270-86.
[http://dx.doi.org/10.1533/9780857090843.2.270]
[19]
Hoffman AS. Hydrogels for biomedical applications. Adv Drug Deliv Rev 2012; 64: 18-23.
[http://dx.doi.org/10.1016/j.addr.2012.09.010] [PMID: 11755703]
[20]
Aizman I, Tate CC, McGrogan M, Case CC. Extracellular matrix produced by bone marrow stromal cells and by their derivative, SB623 cells, supports neural cell growth. J Neurosci Res 2009; 87(14): 3198-206.
[http://dx.doi.org/10.1002/jnr.22146] [PMID: 19530164]
[21]
Dhandayuthapani B, Yoshida Y, Maekawa T, Kumar DS. Polymeric scaffolds in tissue engineering application: A review. Int J Polym Sci 2011; 2011: 1-19.
[http://dx.doi.org/10.1155/2011/290602]
[22]
Vasita R, Katti DS. Nanofibers and their applications in tissue engineering. Int J Nanomedicine 2006; 1(1): 15-30.
[http://dx.doi.org/10.2147/nano.2006.1.1.15] [PMID: 17722259]
[23]
Sokolsky-Papkov M, Agashi K, Olaye A, Shakesheff K, Domb AJ. Polymer carriers for drug delivery in tissue engineering. Adv Drug Deliv Rev 2007; 59(4-5): 187-206.
[http://dx.doi.org/10.1016/j.addr.2007.04.001] [PMID: 17540473]
[24]
Matthews JA, Boland ED, Wnek GE, Simpson DG, Bowlin GL. Electrospinning of collagen type II: A feasibility study. Sage J 2016; 18: 125-34.
[http://dx.doi.org/10.1177/0883911503018002003]
[25]
Anseth KS, Bowman CN, Brannon-Peppas L. Mechanical properties of hydrogels and their experimental determination. Biomaterials 1996; 17(17): 1647-57.
[http://dx.doi.org/10.1016/0142-9612(96)87644-7] [PMID: 8866026]
[26]
Ingber DE. Mechanical signaling and the cellular response to extracellular matrix in angiogenesis and cardiovascular physiology, circulation research. Circ Res 2002; 91(10): 877-87.
[http://dx.doi.org/10.1161/01.RES.0000039537.73816.E5]
[27]
Ryan PL, Foty RA, Kohn J, Steinberg MS. Tissue spreading on implantable substrates is a competitive outcome of cell–cell vs. cell– substratum adhesivity. Proc Natl Acad Sci 2001; 98(8): 4323-7.
[http://dx.doi.org/10.1073/pnas.071615398] [PMID: 11274361]
[28]
Moghe PV, Berthiaume F, Ezzell RM, Toner M, Tompkins RG, Yarmush ML. Culture matrix configuration and composition in the maintenance of hepatocyte polarity and function. Biomaterials 1996; 17(3): 373-85.
[http://dx.doi.org/10.1016/0142-9612(96)85576-1] [PMID: 8745335]
[29]
Baker RW. Membrane technology and applications.Wiley Online Library 2012.
[http://dx.doi.org/10.1002/9781118359686]
[30]
Wang TW, Spector M. Development of hyaluronic acid-based scaffolds for brain tissue engineering. Acta Biomater 2009; 5(7): 2371-84.
[http://dx.doi.org/10.1016/j.actbio.2009.03.033] [PMID: 19403351]
[31]
Venugopal J, Low S, Choon AT, Ramakrishna S. Interaction of cells and nanofiber scaffolds in tissue engineering. J Biomed Mater Res B Appl Biomater 2008; 84B(1): 34-48.
[http://dx.doi.org/10.1002/jbm.b.30841] [PMID: 17477388]
[32]
Barbetta A, Carrino A, Costantini M, Dentini M. Polysaccharide based scaffolds obtained by freezing the external phase of gas-in-liquid foams. Soft Matter 2010; 6(20): 5213-24.
[http://dx.doi.org/10.1039/c0sm00616e]
[33]
Hollister SJ, Levy RA, Chu TM, Halloran JW, Feinberg SE. An image-based approach for designing and manufacturing craniofacial scaffolds. Int J Oral Maxillofac Surg 2000; 29(1): 67-71.
[http://dx.doi.org/10.1034/j.1399-0020.2000.290115.x] [PMID: 10691148]
[34]
Dehghani F, Annabi N, Bornscheuer UT, Khademhosseini A. Engineering porous scaffolds using gas-based techniques. Curr Opin Biotechnol 2011; 22(5): 661-6.
[http://dx.doi.org/10.1016/j.copbio.2011.04.005]
[35]
Quirk RA, France RM, Shakesheff KM, Howdle SM. Supercritical fluid technologies and tissue engineering scaffolds. Curr Opin Solid State Mater Sci 2004; 8(3-4): 313-21.
[http://dx.doi.org/10.1016/j.cossms.2003.12.004]
[36]
Hu X, Liu S, Zhou G, Huang Y, Xie Z, Jing X. Electrospinning of polymeric nanofibers for drug delivery applications. J Control Release 2014; 185: 12-21.
[http://dx.doi.org/10.1016/j.jconrel.2014.04.018] [PMID: 24768792]
[37]
Skardal A, Sarker SF, Crabbé A, Nickerson CA, Prestwich GD. The generation of 3-D tissue models based on hyaluronan hydrogel-coated microcarriers within a rotating wall vessel bioreactor. Biomaterials 2010; 31(32): 8426-35.
[http://dx.doi.org/10.1016/j.biomaterials.2010.07.047] [PMID: 20692703]
[38]
Hasan A, Memic A, Annabi N, et al. Electrospun scaffolds for tissue engineering of vascular grafts. Acta Biomater 2014; 10(1): 11-25.
[http://dx.doi.org/10.1016/j.actbio.2013.08.022] [PMID: 23973391]
[39]
Pant B, Park M, Park SJ. Drug delivery applications of core-sheath nanofibers prepared by coaxial electrospinning: A review. Pharmaceutics 2019; 11(7): 305.
[http://dx.doi.org/10.3390/pharmaceutics11070305] [PMID: 31266186]
[40]
O’Brien FJ, Harley BA, Yannas IV, Gibson LJ. The effect of pore size on cell adhesion in collagen-GAG scaffolds. Biomaterials 2005; 26(4): 433-41.
[http://dx.doi.org/10.1016/j.biomaterials.2004.02.052] [PMID: 15275817]
[41]
Li F, Truong VX, Fisch P, et al. Cartilage tissue formation through assembly of microgels containing mesenchymal stem cells. Acta Biomater 2018; 77: 48-62.
[http://dx.doi.org/10.1016/j.actbio.2018.07.015] [PMID: 30006317]
[42]
Del Bakhshayesh RA, Mostafavi E, Alizadeh E, Asadi N, Akbarzadeh A, Davaran S. Fabrication of three-dimensional scaffolds based on nano-biomimetic collagen hybrid constructs for skin tissue engineering. ACS Omega 2018; 3(8): 8605-11.
[http://dx.doi.org/10.1021/acsomega.8b01219] [PMID: 31458990]
[43]
Tanase CE, Sartoris A, Popa MI, Verestiuc L, Unger RE, Kirkpatrick CJ. In vitro evaluation of biomimetic chitosan–calcium phosphate scaffolds with potential application in bone tissue engineering. Biomed Mater 2013; 8(2): 025002.
[http://dx.doi.org/10.1088/1748-6041/8/2/025002] [PMID: 23343569]
[44]
Da H, Jia SJ, Meng GL, et al. The impact of compact layer in biphasic scaffold on osteochondral tissue engineering. PLoS One 2013; 8(1): e54838.
[http://dx.doi.org/10.1371/journal.pone.0054838] [PMID: 23382984]
[45]
Klimek K, Ginalska G. Proteins and peptides as important modifiers of the polymer scaffolds for tissue engineering applications : A review. Polymers 2020; 12: 844.
[http://dx.doi.org/10.3390/polym12040844]
[46]
Choi SH, Chun SY, Chae SY, et al. Development of a porcine renal extracellular matrix scaffold as a platform for kidney regeneration. J Biomed Mater Res A 2015; 103(4): 1391-403.
[http://dx.doi.org/10.1002/jbm.a.35274] [PMID: 25044751]
[47]
Chan BP, Leong KW. Scaffolding in tissue engineering: General approaches and tissue-specific considerations. Eur Spine J 2008; 17(4): 467-79.
[http://dx.doi.org/10.1007/s00586-008-0745-3] [PMID: 19005702]
[48]
Yazdanian M, Arefi AH, Alam M, et al. Decellularized and biological scaffolds in dental and craniofacial tissue engineering: A comprehensive overview. J mat res technol 2021; 15: 1217-51.
[49]
del Bakhshayesh AR, Annabi N, Khalilov R, et al. Recent advances on biomedical applications of scaffolds in wound healing and dermal tissue engineering. Artif Cells Nanomed Biotechnol 2017; 46(4): 691-705.
[http://dx.doi.org/10.1080/21691401.2017.1349778]
[50]
Pouliot R, Larouche D, Auger FA, et al. Reconstructed human skin produced in vitro and grafted on athymic mice1,2. Transplantation 2002; 73(11): 1751-7.
[http://dx.doi.org/10.1097/00007890-200206150-00010] [PMID: 12084997]
[51]
Singh MR, Saraf S, Vyas A, Jain V, Singh D. Innovative approaches in wound healing: Trajectory and advances. Artif Cells Nanomed Biotechnol 2013; 41: 202-12.
[http://dx.doi.org/10.3109/21691401.2012.716065]
[52]
el Ghalbzouri A, Hensbergen P, Gibbs S, Kempenaar J, van der Schors R, Ponec M. Fibroblasts facilitate re-epithelialization in wounded human skin equivalents. Lab Invest 2004; 84(2003): 102-12.
[http://dx.doi.org/10.1038/labinvest.3700014]
[53]
Falanga V, Margolis D, Alvarez O, et al. Rapid healing of venous ulcers and lack of clinical rejection with an allogeneic cultured human skin equivalent. Arch Dermatol 1998; 134(3): 293-300.
[http://dx.doi.org/10.1001/archderm.134.3.293] [PMID: 9521027]
[54]
Brem H, Balledux J, Bloom T, Kerstein MD, Hollier L. Healing of diabetic foot ulcers and pressure ulcers with human skin equivalent: A new paradigm in wound healing. Arch Surg 2000; 135(6): 627-34.
[http://dx.doi.org/10.1001/archsurg.135.6.627] [PMID: 10843357]
[55]
Velnar T, Gradisnik L. Tissue augmentation in wound healing: The role of endothelial and epithelial cells. Med Arch 2018; 72: 444-8.
[http://dx.doi.org/10.5455/medarh.2018.72.444-448]
[56]
Strecker-McGraw MK, Jones TR, Baer DG. Soft tissue wounds and principles of healing. Emerg Med Clin North Am 2007; 25(1): 1-22.
[http://dx.doi.org/10.1016/j.emc.2006.12.002] [PMID: 17400070]
[57]
Dias AMA, Braga MEM, Seabra IJ, Ferreira P, Gil MH, de Sousa HC. Development of natural-based wound dressings impregnated with bioactive compounds and using supercritical carbon dioxide. Int J Pharm 2011; 408(1-2): 9-19.
[http://dx.doi.org/10.1016/j.ijpharm.2011.01.063] [PMID: 21316432]
[58]
Ramshaw JAM, Peng YY, Glattauer V, Werkmeister JA. Collagens as biomaterials. J Mat Sci Mat Med 2008; 20(1.20): 3-8.
[http://dx.doi.org/10.1007/s10856-008-3415-4]
[59]
Doillon CJ, Silver FH. Collagen-based wound dressing: Effects of hyaluronic acid and firponectin on wound healing. Biomaterials 1986; 7(1): 3-8.
[http://dx.doi.org/10.1016/0142-9612(86)90080-3] [PMID: 3955155]
[60]
Prus-Walendziak W, Kozlowska J. Lyophilized emulsions in the form of 3d porous matrices as a novel material for topical application. Materials 2021; 14(4): 950.
[http://dx.doi.org/10.3390/ma14040950] [PMID: 33671458]
[61]
Ishihara M, Nakanishi K, Ono K, et al. Photocrosslinkable chitosan as a dressing for wound occlusion and accelerator in healing process. Biomaterials 2002; 23(3): 833-40.
[http://dx.doi.org/10.1016/S0142-9612(01)00189-2] [PMID: 11771703]
[62]
Sill TJ, von Recum HA. Electrospinning: Applications in drug delivery and tissue engineering. Biomaterials 2008; 29(13): 1989-2006.
[http://dx.doi.org/10.1016/j.biomaterials.2008.01.011] [PMID: 18281090]
[63]
Ranganath SH, Wang CH. Biodegradable microfiber implants delivering paclitaxel for post-surgical chemotherapy against malignant glioma. Biomaterials 2008; 29(20): 2996-3003.
[http://dx.doi.org/10.1016/j.biomaterials.2008.04.002] [PMID: 18423584]
[64]
Ali MA, Mondal K, Singh C, Malhotra DB, Sharma A. Anti-epidermal growth factor receptor conjugated mesoporous zinc oxide nanofibers for breast cancer diagnostics. Nanoscale 2015; 7(16): 7234-45.
[http://dx.doi.org/10.1039/C5NR00194C] [PMID: 25811908]
[65]
Wang Z, Qian Y, Li L, et al. Evaluation of emulsion electrospun polycaprolactone/hyaluronan/epidermal growth factor nanofibrous scaffolds for wound healing. J Biomater Appl 2015; 30(6): 686-98.
[http://dx.doi.org/10.1177/0885328215586907]
[66]
Fiume L, Baglioni M, Bolondi L, Farina C, Di Stefano G. Doxorubicin coupled to lactosaminated human albumin: A hepatocellular carcinoma targeted drug. Drug Discov Today 2008; 13(21-22): 1002-9.
[http://dx.doi.org/10.1016/j.drudis.2008.07.009] [PMID: 18755287]
[67]
Sampath M, Lakra R, Korrapati P, Sengottuvelan B. Curcumin loaded poly (lactic-co-glycolic) acid nanofiber for the treatment of carcinoma. Colloids Surf B Biointerfaces 2014; 117: 128-34.
[http://dx.doi.org/10.1016/j.colsurfb.2014.02.020] [PMID: 24646452]
[68]
Hinderer S, Schesny M, Bayrak A, et al. Engineering of fibrillar decorin matrices for a tissue-engineered trachea. Biomaterials 2012; 33(21): 5259-66.
[http://dx.doi.org/10.1016/j.biomaterials.2012.03.075] [PMID: 22521489]
[69]
Schaefer L, Schaefer RM. Proteoglycans: From structural compounds to signaling molecules. Cell Tissue Res 2010; 339(1): 237-46.
[http://dx.doi.org/10.1007/s00441-009-0821-y] [PMID: 19513755]
[70]
Agarwal S, Wendorff JH, Greiner A. Use of electrospinning technique for biomedical applications. Polymer 2008; 49(26): 5603-21.
[http://dx.doi.org/10.1016/j.polymer.2008.09.014]
[71]
Tong HW, Mutlu BR, Wackett LP, Aksan A. Manufacturing of bioreactive nanofibers for bioremediation. Biotechnol Bioeng 2014; 111(8): 1483-93.
[http://dx.doi.org/10.1002/bit.25208] [PMID: 24615064]
[72]
Mangır N, Bullock AJ, Roman S, Osman N, Chapple C, MacNeil S. Production of ascorbic acid releasing biomaterials for pelvic floor repair. Acta Biomater 2016; 29: 188-97.
[http://dx.doi.org/10.1016/j.actbio.2015.10.019] [PMID: 26478470]
[73]
Hinderer S, Layland SL, Schenke-Layland K. ECM and ECM-like materials: Biomaterials for applications in regenerative medicine and cancer therapy. Adv Drug Deliv Rev 2016; 97: 260-9.
[http://dx.doi.org/10.1016/j.addr.2015.11.019] [PMID: 26658243]
[74]
Yang G, Wang J, Wang Y, Li L, Guo X, Zhou S. An implantable active-targeting micelle-in-nanofiber device for efficient and safe cancer therapy. ACS Nano 2015; 9(2): 1161-74.
[http://dx.doi.org/10.1021/nn504573u] [PMID: 25602381]
[75]
Langer R, Vacanti JP. Tissue engineering. Science 1993; 260(5110): 920-6.
[http://dx.doi.org/10.1126/science.8493529] [PMID: 8493529]
[76]
Niklason LE, Langer R. Prospects for organ and tissue replacement. JAMA 2001; 285(5): 573-6.
[http://dx.doi.org/10.1001/jama.285.5.573] [PMID: 11176861]
[77]
Chevalier E, Chulia D, Pouget C, Viana M. Fabrication of porous substrates: A review of processes using pore forming agents in the biomaterial field. J Pharm Sci 2008; 97(3): 1135-54.
[http://dx.doi.org/10.1002/jps.21059] [PMID: 17688274]
[78]
Yang S, Leong KF, Du Z, Chua CK. The design of scaffolds for use in tissue engineering. Part II. Rapid prototyping techniques. Tissue Eng 2002; 8(1): 1-11.
[http://dx.doi.org/10.1089/107632702753503009] [PMID: 11886649]
[79]
Hollister SJ. Porous scaffold design for tissue engineering. Nat Mater 2005; 4(7): 518-24.
[http://dx.doi.org/10.1038/nmat1421] [PMID: 16003400]
[80]
Hutmacher DW, Sittinger M, Risbud MV. Scaffold-based tissue engineering: Rationale for computer-aided design and solid free- form fabrication systems. Trends Biotechnol 2004; 22(7): 354-62.
[http://dx.doi.org/10.1016/j.tibtech.2004.05.005] [PMID: 15245908]
[81]
Dhariwala B, Hunt E, Boland T. Rapid prototyping of tissue-engineering constructs, using photopolymerizable hydrogels and stereolithography. Tissue Eng 2004; 10(9-10): 1316-22.
[http://dx.doi.org/10.1089/ten.2004.10.1316] [PMID: 15588392]
[82]
Perry TE, Roth SJ. Cardiovascular tissue engineering: Constructing living tissue cardiac valves and blood vessels using bone marrow, umbilical cord blood, and peripheral blood cells. J Cardiovasc Nurs 2003; 18(1): 30-7.
[http://dx.doi.org/10.1097/00005082-200301000-00005] [PMID: 12537087]
[83]
Twal WO, Klatt SC, Harikrishnan K, et al. Cellularized microcarriers as adhesive building blocks for fabrication of tubular tissue constructs. Ann Biomed Eng 2014; 42(7): 1470-81.
[http://dx.doi.org/10.1007/s10439-013-0883-6] [PMID: 23943070]
[84]
Hall S. Axonal regeneration through acellular muscle grafts. J Anat 1997; 190(1): 57-71.
[http://dx.doi.org/10.1046/j.1469-7580.1997.19010057.x] [PMID: 9034882]
[85]
Badylak SF. Xenogeneic extracellular matrix as a scaffold for tissue reconstruction. Transpl Immunol 2004; 12(3-4): 367-77.
[http://dx.doi.org/10.1016/j.trim.2003.12.016] [PMID: 15157928]
[86]
Gilbert T, Sellaro T, Badylak S. Decellularization of tissues and organs. Biomaterials 2006; 27(19): 3675-83.
[http://dx.doi.org/10.1016/j.biomaterials.2006.02.014] [PMID: 16519932]
[87]
Schmidt CE, Baier JM. Acellular vascular tissues: Natural biomaterials for tissue repair and tissue engineering. Biomaterials 2000; 21(22): 2215-31.
[http://dx.doi.org/10.1016/S0142-9612(00)00148-4] [PMID: 11026628]
[88]
Takezawa T, Mori Y, Yoshizato K. Cell culture on a thermo-responsive polymer surface. Biotechnology 1990; 8(9): 854-6.
[http://dx.doi.org/10.1038/nbt0990-854]
[89]
Okano T, Yamada N, Sakai H, Sakurai Y. A novel recovery system for cultured cells using plasma-treated polystyrene dishes grafted with poly(N-isopropylacrylamide). J Biomed Mater Res 1993; 27(10): 1243-51.
[http://dx.doi.org/10.1002/jbm.820271005] [PMID: 8245039]
[90]
Okano T, Yamada N, Okuhara M, Sakai H, Sakurai Y. Mechanism of cell detachment from temperature-modulated, hydrophilic-hydrophobic polymer surfaces. Biomaterials 1995; 16(4): 297-303.
[http://dx.doi.org/10.1016/0142-9612(95)93257-E] [PMID: 7772669]
[91]
Nishida K, Yamato M, Hayashida Y, et al. Corneal reconstruction with tissue-engineered cell sheets composed of autologous oral mucosal epithelium. N Engl J Med 2004; 351(12): 1187-96.
[http://dx.doi.org/10.1056/NEJMoa040455] [PMID: 15371576]
[92]
Shimizu T, Sekine H, Yamato M, Okano T. Cell sheet-based myocardial tissue engineering: new hope for damaged heart rescue. Curr Pharm Des 2009; 15(24): 2807-14.
[http://dx.doi.org/10.2174/138161209788923822] [PMID: 19689351]
[93]
Jiyoung MD, Kam WL. Myogenic induction of aligned mesenchymal stem cell sheets by culture on thermally responsive electrospun nanofibers. Adv Mater 2007; 19: 2775-9.
[http://dx.doi.org/10.1002/adma.200602159]
[94]
Isenberg BC, Tsuda Y, Williams C, et al. A thermoresponsive, microtextured substrate for cell sheet engineering with defined structural organization. Biomaterials 2008; 29(17): 2565-72.
[http://dx.doi.org/10.1016/j.biomaterials.2008.02.023] [PMID: 18377979]
[95]
da Silva RMP, Mano JF, Reis RL. Smart thermoresponsive coatings and surfaces for tissue engineering: Switching cell-material boundaries. Trends Biotechnol 2007; 25(12): 577-83.
[http://dx.doi.org/10.1016/j.tibtech.2007.08.014] [PMID: 17997178]
[96]
Leucht P, Lee S, Yim N. Wnt signaling and bone regeneration: Can’t have one without the other. Biomaterials 2019; 196: 46-50.
[http://dx.doi.org/10.1016/j.biomaterials.2018.03.029] [PMID: 29573821]
[97]
Boerckel JD, Kolambkar YM, Stevens HY, Lin ASP, Dupont KM, Guldberg RE. Effects of in vivo mechanical loading on large bone defect regeneration. J Orthop Res 2012; 30(7): 1067-75.
[http://dx.doi.org/10.1002/jor.22042] [PMID: 22170172]
[98]
Mohammadi M, Alibolandi M, Abnous K, Salmasi Z, Jaafari MR, Ramezani M. Fabrication of hybrid scaffold based on hydroxyapatite-biodegradable nanofibers incorporated with liposomal formulation of BMP-2 peptide for bone tissue engineering. Nanomedicine 2018; 14(7): 1987-97.
[http://dx.doi.org/10.1016/j.nano.2018.06.001] [PMID: 29933024]
[99]
Sun X, Wu Z, He D, et al. Bioactive injectable polymethylmethacrylate/silicate bioceramic hybrid cements for percutaneous vertebroplasty and kyphoplasty. J Mech Behav Biomed Mater 2019; 96: 125-35.
[http://dx.doi.org/10.1016/j.jmbbm.2019.04.044] [PMID: 31035063]
[100]
Ustek S, Kismet K, Akkus MA, Ozcan AH, Aydogan A, Renda N. Effect of povidone-iodine liposome hydrogel on colonic anastomosis. Eur Surg Res 2005; 37(4): 242-5.
[http://dx.doi.org/10.1159/000087870]
[101]
Contents: (Adv. Healthcare Mater. 19/2017). In: Advanced Healthcare Materials 2017; 6(19).
[http://dx.doi.org/10.1002/adhm.201770100]
[102]
Hurler J, Sørensen KK, Fallarero A, Vuorela P, Škalko-Basnet N. Liposomes-in-hydrogel delivery system with mupirocin: in vitro antibiofilm studies and in vivo evaluation in mice burn model. BioMed Res Int 2013; 2013: 1-8.
[http://dx.doi.org/10.1155/2013/498485] [PMID: 24369533]
[103]
Ziegler G, Grabher P, Thompson A, et al. Progressive neurodegeneration following spinal cord injury. Neurology 2018; 90(14): e1257-66.
[http://dx.doi.org/10.1212/WNL.0000000000005258] [PMID: 29514946]
[104]
Li X, Dai J. Bridging the gap with functional collagen scaffolds: Tuning endogenous neural stem cells for severe spinal cord injury repair. Biomater Sci 2018; 6(2): 265-71.
[http://dx.doi.org/10.1039/C7BM00974G] [PMID: 29265131]
[105]
Zhao Y, Xiao Z, Chen B, Dai J. The neuronal differentiation microenvironment is essential for spinal cord injury repair. Organogenesis 2017; 13(3): 63-70.
[http://dx.doi.org/10.1080/15476278.2017.1329789] [PMID: 28598297]
[106]
Melling GE, Colombo JS, Avery SJ, et al. Liposomal delivery of demineralized dentin matrix for dental tissue regeneration. Tissue Eng Part A 2018; 24(13-14): 1057-65.
[http://dx.doi.org/10.1089/ten.tea.2017.0419] [PMID: 29316874]
[107]
Zajda J, Farag F. Urolastic-a new bulking agent for the treatment of women with stress urinary incontinence: Outcome of 12 months follow up. Adv Urol 2013; 2013: 1-5.
[http://dx.doi.org/10.1155/2013/724082] [PMID: 24454351]
[108]
Jung S, Oh H-K, Kim M-S, Lee K-Y, Park H, Kook M-S. Effect of gellan gum/tuna skin film in guided bone regeneration in artificial bone defect in rabbit calvaria. Materials 2020; 13(6): 1318.
[http://dx.doi.org/10.3390/ma13061318]
[109]
Kovačević J, Prucková Z, Pospíšil T, Kašpárková V, Rouchal M, Vícha R. A new hyaluronan modified with β-cyclodextrin on hydroxymethyl groups forms a dynamic supramolecular network. Molecules 2019; 24(21): 3849.
[http://dx.doi.org/10.3390/molecules24213849]
[110]
Seidlits SK, Drinnan CT, Petersen RR, Shear JB, Suggs LJ, Schmidt CE. Fibronectin–hyaluronic acid composite hydrogels for three-dimensional endothelial cell culture. Acta Biomater 2011; 7(6): 2401-9.
[http://dx.doi.org/10.1016/j.actbio.2011.03.024] [PMID: 21439409]
[111]
Erickson IE, Kestle SR, Zellars KH, et al. High mesenchymal stem cell seeding densities in hyaluronic acid hydrogels produce engineered cartilage with native tissue properties. Acta Biomater 2012; 8(8): 3027-34.
[http://dx.doi.org/10.1016/j.actbio.2012.04.033] [PMID: 22546516]
[112]
Kim HJ, Kim KK, Park IK, Choi BS, Kim JH, Kim MS. Hybrid scaffolds composed of hyaluronic acid and collagen for cartilage regeneration. Tiss Eng Regen Med 2012; 9: 57-62.
[http://dx.doi.org/10.1007/s13770-012-0007-7]
[113]
Guo Y, Yuan T, Xiao Z, et al. Hydrogels of collagen/chondroitin sulfate/hyaluronan interpenetrating polymer network for cartilage tissue engineering. J Mater Sci Mater Med 2012; 23(9): 2267-79.
[http://dx.doi.org/10.1007/s10856-012-4684-5]
[114]
Wang X, He J, Wang Y, Cui FZ. Hyaluronic acid-based scaffold for central neural tissue engineering. Interface Focus 2012; 2(3): 278-91.
[http://dx.doi.org/10.1098/rsfs.2012.0016] [PMID: 23741606]
[115]
Zhong J, Chan A, Morad L, Kornblum HI, Guoping Fan , Carmichael ST. Hydrogel matrix to support stem cell survival after brain transplantation in stroke. Neurorehabil Neural Repair 2010; 24(7): 636-44.
[http://dx.doi.org/10.1177/1545968310361958] [PMID: 20424193]
[116]
Nesti LJ, Li W-J, Shanti RM, et al. Intervertebral disc tissue engineering using a novel hyaluronic acid–nanofibrous scaffold (hanfs) amalgam. Tiss Eng Part A 2008; 14: 1527-37. Available from: https://www.academia.edu/11443224/Intervertebral_Disc_Tissue_Engineering_Using_a_Novel_Hyaluronic_Acid_Nanofibrous_Scaffold_HANFS_Amalgam
[117]
Burdick JA, Prestwich GD. Hyaluronic acid hydrogels for biomedical applications. Adv Mater 2011; 23(12): H41-56.
[http://dx.doi.org/10.1002/adma.201003963]
[118]
Ekaputra AK, Prestwich GD, Cool SM, Hutmacher DW. The three-dimensional vascularization of growth factor-releasing hybrid scaffold of poly (ɛ-caprolactone)/collagen fibers and hyaluronic acid hydrogel. Biomaterials 2011; 32(32): 8108-17.
[http://dx.doi.org/10.1016/j.biomaterials.2011.07.022] [PMID: 21807407]
[119]
Ma J, Holden K, Zhu J, Pan H, Li Y. The application of three-dimensional collagen-scaffolds seeded with myoblasts to repair skeletal muscle defects. J Biomed Biotechnol 2011; 2011: 812135.
[http://dx.doi.org/10.1155/2011/812135]
[120]
Detamore, Decellularized hyaline cartilage powder for tissue scaffolds. US10722614B2, 2014.
[121]
Seliktar D, Almany L. Pegylated fibrinogen precursor molecule. US9474830B2, 2015.
[122]
Self-assembling biomimetic hydrogels having bioadhesive properties. US9295761B2, Available from: https://patents.google.com/patent/US9295761B2/en (accessed September 1, 2021).
[123]
Rockwood DN, Preda RC, Yucel T, Wang X, Lovett ML, Kaplan DL. Materials fabrication from Bombyx mori silk fibroin. Nat Protoc 2013; 6(10): 1612-31.
[124]
Arinzeh, Scaffold for tissue growth and repair. Biochem Soc Trans 2016; 9: 2939-47.
[125]
Pina S, Ribeiro VP, Paiva OC, Correlo VM, Oliveira JM, Reis RL. Tissue engineering scaffolds: Future perspectives. Handbook of tissue engineering scaffolds: Volume one. Woodhead Publishing 2019; pp. 165-85.
[http://dx.doi.org/10.1016/B978-0-08-102563-5.00009-5]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy