1. Trang chủ
  2. » Giáo Dục - Đào Tạo

INVESTIGATING THE COMPATIBILITY OF a PROTOTYPE SCAFFOLD IN a NOVEL NEAR CELLSHEET APPROACH

135 294 0

Đang tải... (xem toàn văn)

Tài liệu hạn chế xem trước, để xem đầy đủ mời bạn chọn Tải xuống

THÔNG TIN TÀI LIỆU

Nội dung

ENGINEERING PCL-BASED NANO-MATERIAL FOR TISSUE REPAIR ALEX FOO HSIEN LOONG B.Eng. (Hons.), NUS A THESIS SUBMITTED FOR THE DEGREE OF DOCTOR OF PHILOSOPHY NUS FACULTY OF MEDICINE GRADUATE PROGRAM IN BIOENGINEERING NATIONAL UNIVERSITY OF SINGAPORE 2009 ACKNOWLEDGEMENTS I am greatly indebted to the following people in the course of this project. I will like to thank my thesis supervisors Prof Teoh Swee Hin and Prof Hanry Yu for their guidance in this project. I will also like to thank my project collaborator Dr Lou Yan-Ru for her generous support in hMSC culture, in particularly her patience and clear advice with various molecular biotechnologies such as RT-PCR and Western Blotting. I will like to thank Dr Jeremy Teo for passing down his extensive knowledge in microCT imaging and his gallant help to analyze the results. I will also like to specially acknowledge my most under-appreciated cinematographer Ms Ong Siew Min for her assistance during video filming. I will like to thank the following people for their friendship during my stay: Mr Talha Arooz, Dr Suzanne Ng San San, Dr Khong Yuet Mei, Mr Lim Teck Chuan, Dr Edwin Chow, Dr Kan Shyi-Herng, Dr Amy Chou and Dr Toh Xue Li. I will like to thank Prof Jackie Ying, Ms Noreena AbuBakar, IBN administrative, safety and facilities department for their support in providing me a pleasant environment to work in. I will like to acknowledge the research funding from IBN, BMRC, A-Star, as well as the generous scholarship from NUS I will like to thank my family for being understanding, patient and supportive of my own selfish pursuits. I will also like to humbly dedicate this thesis to my niece Sydney Leong and hope that this will inspire her in her future endeavors when she grows up. Last but not least, I will like to thank God for His unfailing love during this journey of faith. TABLE OF CONTENTS ACKNOWLEDGEMENTS --------------------------------------------------------------- SUMMARY ---------------------------------------------------------------------------------- LIST OF PUBLICATIONS -------------------------------------------------------------- LIST OF TABLES -------------------------------------------------------------------------- LIST OF FIGURES ------------------------------------------------------------------------ LIST OF VIDEOS ------------------------------------------------------------------------- 12 LIST OF SYMBOLS AND ABBREVIATIONS ------------------------------------- 13 I INTRODUCTION ---------------------------------------------------------------- 17 II LITERATURE REVIEW ------------------------------------------------------- 22 2.1 The use of scaffolds in TE ----------------------------------------------- 22 2.2. Nanofibers ----------------------------------------------------------------- 24 2.2.1 Non-conventional fabrication approach ---------------------- 25 2.2.2 Polyelectrolyte complexation ---------------------------------- 26 2.2.3 Self-assembly ----------------------------------------------------- 28 2.2.4 Electrospinning --------------------------------------------------- 32 III OBJECTIVES --------------------------------------------------------------------- 40 IV MATERIALS AND METHODS ----------------------------------------------- 43 4.1 Fabrication of FSS -------------------------------------------------------- 43 V 4.2 Characterization of FSS ------------------------------------------------- 44 4.3 Non-invasive surface modification of FSS ---------------------------- 47 4.4 Cell culture ----------------------------------------------------------------- 49 4.5 Biological characterizations --------------------------------------------- 50 4.6 Specific tests for primary hepatocytes --------------------------------- 51 4.7 Specific tests for osteoblastic hMSC ----------------------------------- 52 4.8 Statistical methods -------------------------------------------------------- 55 RESULTS --------------------------------------------------------------------------- 56 5.1 Specific Aim ------------------------------------------------------------- 56 5.2 Specific Aim ------------------------------------------------------------- 76 5.3 Specific Aim ------------------------------------------------------------- 89 VI DISCUSSION ---------------------------------------------------------------------- 110 VII CONCLUSIONS ------------------------------------------------------------------ 117 VIII REFERENCES -------------------------------------------------------------------- 118 SUMMARY Conventional approach to TE commonly involves the use of synthetic scaffoldings that often lack the nano-scale features typically found in the native environment. Such dissimilarity has been speculated to be one of the instrumental factors contributing to the poor maintenance of the cells under in vitro conditions. An alternative solution is to culture them on fibrillar surfaces generated rapidly from emerging technologies such as electrospinning. Our group has come up with a new PCL-based nano-material design (FSS) that can ensure the structural stability of the otherwise fragile substrate irregardless of the pore dimensions. A highly permeable material with ease of handling can therefore be fabricated and its potential use as a sandwiching or stacking membrane was examined. Morphological and functionality studies on overlaid primary hepatocytes revealed a 3D-like cytoskeletal structure and improved urea secretion. A more extensive bone differentiation profile was also observed in hMSC when individual pre-seeded low density FSS were stacked together prior to their exposure to osteogenic medium. Preliminary results also identified physical communications to play a significant role in priming the cells to be more receptive to external signaling cues. Such benefits in biological responses were however not extended to hepatic layers probably due to an inadequate cell density used during the test. We propose here an alternative material that can be stacked up to better mimic the stratified architecture of the innate surrounding. LIST OF PUBLICATIONS (1) Foo HL, Taniguchi A, Yu H, Okano T, Teoh SH, Catalytic Surface Modification of Rolled-Milled Poly (ε-Caprolactone) Biaxially Stretched to Ultra-Thin Dimension. Materials Science & Engineering C – Biomimetic and Supramolecular Systems, 2007. 27 (2): p. 299 – 303. (2) Ong SM, Zhang C, Toh YC, Kim SH, Foo HL, Tan CH, van Noort D, Park S, Yu H, A Gel-Free 3D Microfluidic Cell Culture System. Biomaterials, 29 (22): p 3237 – 3244. LIST OF PATENTS (1) Foo A, Yu H, Bioactive Scaffold With Controllable Mass Transfer Properties Independent of Mechanical Strength. Provisional Patent (25 June 2008). LIST OF TABLES Table Current status in TE [Stock UA, 2001] 21 Table Desirable features in an ideal scaffold [Murugan R, 2007] 24 Table Mechanical properties of thin films from various processes [Tiaw KS, 2007]. 58 Table Stiffness in various substrates 60 Table Comparing the fluorescence density between the top and bottom layer 64 Table Comparing XPS stoichometric ratios between various surfaces 71 Table Comparing XPS stoichometric ratios between surfaces anchored or grafted with collagen 72 Table Comparing the water permeability between different substrates 80 Table Urea response towards change in fiber density and overlay time 87 Table 10 Expression of bone-specific genes with respect to stacking 96 Table 11 Gene and protein expression with respect to overlay time 99 LIST OF FIGURES Figure UNOS organ transplant statistics for 1990 to 1999 documenting the wait-listed patients (O) and transplants (●) [Drury JL, 2003] 18 Figure Biaxially stretched PCL film and its properties 58 Figure Fabrication of FSS and its morphology 59 Figure Improved mechanical strength with FSS 60 Figure Improved ease of harvesting with FSS 62 Figure Non-invasive surface modification of FSS via collagen gelation and anchoring 64 Figure Non-invasive surface modification with Fe (II) catalyst via PAAc-intermediate 66 Figure Varying several experimental conditions to optimize the PAAc density on surface 69 Figure Detecting collagen on PAAc-grafted FSS 71 Figure 10 Comparing between surfaces previously anchored or grafted with collagen 72 Figure 11 Investigating cell behavior on modified high density FSS 73 Figure 12 Investigating cell behavior on modified low density FSS 75 Figure 13 Benefits of membrane-based sandwich model in liver TE 78 Figure 14 Effect of FSS on mass transfer 80 Figure 15 Effect of FSS on viability of primary hepatocytes during overlay 81 Figure 16 Investigating the benefits of sandwiching primary hepatocytes with FSS 83 Figure 17 Optimization of sandwiched culture with FSS 85 Figure 18 Increasing the transplanted cell number per unit area 91 Figure 19 Improvements in osteogenic responses of hMSC during stacking 94 Figure 20 Examining the effect of fiber density on stacking consequences in hMSC 99 Figure 21 The presence and importance of physical contacts in stacking configuration 100 Figure 22 Establishing sparse and dense cultures with the same initial seeding number 102 Figure 23 Comparing the gene expression of hMSC post-exposed to vitamin D3-supplemented osteogenic medium in dense and dispersed culture 103 Figure 24 Total and normalized functions of primary hepatocytes in a nonstacked and stacked configuration. 106 Figure 25 Investigating the effect of fiber density on primary hepatocyte during stacking. 107 Figure 26 Investigating the presence and importance of physical contacts in stacking model of primary hepatocytes 108 Figure 27 Investigating the reason for stacked primary hepatocytes to exhibit inferior functions on low density FSS in spite of the presence of physical contacts 108 10 Dalby MJ. (2005). Medical engineering & physics. Topographically induced direct cell mechanotransduction. 27: 730 – 742 Dang JM, Leong KW. (2007). Advanced materials. Myogenic induction of aligned mesenchymal stem cell sheets by culture on thermally responsive electrospun nanofibers. 19: 2775 – 2779 Dar A, Shachar M, Leor J, Cohen S. (2002). Biotechnology and bioengineering. Optimization of cardiac cell seeding and distribution in 3D porous alginate scaffolds. 80: 305 – 312 Davis ME, Heish PCH, Takahashi T, Song Q, Zhang SG, Kamm RD, Grodzinsky AJ, Anversa P, Lee RT. (2006). Proceedings of the National Academy of Sciences of the United States of America. Local myocardial insulin-like growth factor (IGF-1) delivery with biotinylated peptide nanofibers improves cell therapy for myocardial infarction. 103 (21): 8155 – 8160 Deitzel JM, Kleinmeyer J, Harris D, Tan NCB. (2001). Polymer. The effect of processing variable on the morphology of electrospun nanofibers and textiles. 42: 261 – 272 Demir MM, Yilgor I, Yilgor E, Erman B. (2002). Polymer. Electrospinning of polyurethane fibers. 43: 3303 – 3309 Deng C, Ueda E, Chen KE, Bula C, Norman AW, Luben RA, Walker AM. (2009). Molecular endocrinology. Prolactin blocks nuclear translocation of VDR by regulating its interaction with BRCA1 in osteosarcoma cells. 23: 226 - 236 Drury JL, Mooney DJ. (2003). Biomaterials. Hydrogels for tissue engineering: scaffold design variables and applications. 24: 4337 – 4351 Du Y, Han RB, Wen F, Ng SSS, Xia L, Wohland T, Leo HL, Yu H. (2008). Biomaterials. Synthetic sandwich culture of 3D hepatocyte monolayer. 29 (3): 290 – 301 Dunn JCY, Tompkins RG, Yarmush ML. (1991). Biotechnology progress. Long-term in vitro function of adult hepatocytes in a collagen sandwich configuration. 7: 237 – 245 120 Dvir-Ginzberg M, Gamlieli-Bonshtein I, Agbaria R, Cohen S. (2003). Tissue engineering. Liver tissue engineering within alginate scaffolds: effects of cell-seeding density on hepatocyte viability, morphology and function. 9(4): 757 – 766 Eda G, Shivkumar S. (2007). Journal of applied polymer science. Bead-to-fiber transition in electrospun polystyrene. 106: 475 – 487 Eichhorn SJ, Sampson WW. (2005). Journal of the royal society interface. Statistical geometry of pores and statistics of porous nanofibrous assemblies. 2: 309 Elias KL, Price RL, Webster TJ. (2002). Biomaterials. Enhanced functions of osteoblasts on nanometer diameter carbon fibers. 23: 3279 Etheridge SL, Spencer GJ, Heath DJ, Genever PG. (2004). Stem cells. Expression profiling and functional analysis of Wnt signaling mechanisms in mesenchymal stem cells. 22: 849 – 860 Fang X, Reneker DH. (1997). Journal of macromoleular science, part b: physics. DNA fibers by electrospinning. 36 (2): 169 – 173 Feng ZQ, Chu XH, Huang NP, Wang T, Wang YC, Shi XL, Ding YT, Gu ZZ. (2009). Biomaterials. The effect of nanofibrous galactosylated chitosan scaffolds on the formation of rat primary hepatocyte aggregates and the maintenance of liver function. 30 (14): 2753 – 2763 Foo HL, Taniguchi A, Yu H, Okano T, Teoh SH. (2007). Material science and engineering c. Catalytic surface modification of roll-milled poly(ε-caprolactone) biaxially stretched to ultra-thin dimension. 27: 299 – 303 Fretz JA, Zella LA, Kim ST, Shevde NK, Wesley Pike J. (2007). Journal of steroid biochemistry & molecular biology. 1, 25-dihydroxyvitamin D3 induces expression of the Wnt signaling co-regulator LRP5 via regulatory elements located significantly downstream of the gene’s transcriptional start site. 103: 440 – 445 Garreta E, Genove E, Borros S, Semino CE. (2006). Tissue engineering. Osteogenic differentiation of mouse embryonic stem cells and mouse embryonic fibroblasts in a three-dimensional self-assembling peptide scaffold. 12 (8): 2215 – 2227 121 Gelain F, Horii A, Zhang SG. (2007). Macromolecular bioscience. Designer selfassembling peptide scaffolds for 3D tissue cell cultures and regenerative medicine. 7: 544 – 551 Geng XY, Kwon OH, Jang JH. (2005). Biomaterials. Electrospinning of chitosan dissolved in concentrated acetic acid solution. 26: 5427 – 5432 Genove E, Shen C, Zhang SG, Semino CE. (2005). Biomaterials. The effect of functionalized self-assembling peptide scaffolds on human aortic endothelial cell function. 26: 3341 – 3351 Goldstein AS. (2001). Tissue engineering. Effect of seeding osteoprogenitor cells as dense clusters on cell growth and differentiation. 7(6): 817 – 827 Grafahrend D, Calvet JL, Salber J, Dalton PD, Moeller M, Klee D. (2007). Journal of materials science: materials in medicine. Biofunctionalized poly(ethylene glycol)block-poly(ε-caprolactone) nanofibers for tissue engineering. Gupta B, Plummer C, Bisson I, Frey P, Hilborn J. (2002). Biomaterials. Plasmainduced graft polymerization of acrylic acid onto poly (ethylene terephthalate) films: characterization and human smooth muscle cell growth on grafted films. 23: 863 Gupta P, Elkins C, Long TE, Wilkes GL. (2005). Polymer. Electrospinning of linear homopolymers of poly (methyl methacrylate): exploring relationships between fiber formation, viscosity, molecular weight and concentration in a good solvent. 46: 4799 – 4810 Hamilton GA, Westmorel C, George AE. (2001). In vitro cellular & developmental biology, animal. Effects of medium composition on the morphology and function of rat hepatocytes cultured as spheroids and monolayers. 37 (10): 656 – 667 Hartgerink JD, Beniash E, Stupp SL. (2001). Science. Self-assembly and mineralization of peptide-amphiphile nanofibers. 294: 1684 – 1688 Hartig SM, Greene RR, Dikov MM, Prokop A, Davidson JM. (2007). Pharmaceutical research. Multifunctional nanoparticulate polyelectrolyte complexes. 24 (12): 2353 – 2369 122 Hasegawa M, Yamato M, Kikuchi A, Okano T, Ishikawa I. (2005). Tissue engineering. Human periodontal ligament cell sheets can regenerate periodontal ligament tissue in an athymic rat model. 11 (3/4): 469 – 478 He CC, Gu ZY. (2003). Journal of applied polymer science. Studies on acrylic acidgrafted polyester fabrics by electron beam preirradiation method. I. Effects of process parameters on graft ratio and characterization of grafting products. 89: 3931 Hollister SJ. (2005). Nature materials. Porous scaffold design for tissue engineering. 4: 518 – 524 Holy CE, Shoichet MS, Davies JE. (2000). Journal of biomedical materials research. Engineering three-dimensional bone tissue in vitro using biodegradable scaffolds: investigating initial cell-seeding density and culture period. 51: 376 - 382 Hoshiba T, Nagahara H, Cho CS, Tagawa YI, Akaike T. (2007). Biomaterials. Primary hepatocyte survival on non-integrin-recognizable matrices without the activation of Akt signaling. 28: 1093 – 1104 Htay AS, Teoh SH, Hutmacher DW. (2004). Journal of biomaterials science, polymer edition. Development of perforated microthin poly(ε-caprolactone) films as matrices for membrane tissue engineering. 15 (5): 683 – 700 http://ustransplant.org Hu W, Yim EKF, Reano RM, Leong KW, Pang SW. (2005). Journal of vacuum science and technology B. Effects of nanoimprinted patterns in tissue-culture polystyrene on cell behavior. 23 (6): 2984 – 2989 Hui TY, Cheung KMC, Cheung WL, Chan D, Chan BP. (2008). Biomaterials. In vitro chondrogenic differentiation of human mesenchymal stem cells in collagen microspheres: influence of cell seeding density and collagen concentration. 29: 3201 – 3212. Hutmacher DW, Sittinger M, Risbud MV. (2004). Trends in biotechnology. Scaffoldbased tissue engineering: rationale for computer-aided design and solid free-form fabrication systems. 22 (7): 354 – 362 123 Hutmacher DW. (2001). Journal of biomaterials science: polymer edition. Scaffold design and fabrication technologies for engineering tissues – state of the art and future perspectives. 12 (1): 107 – 124 Hutmacher DW. (2006). Expert review in medical devices. Regenerative medicine will impact, but not replace, the medical device industry. 3(4): 409 – 412 Ishii O, Shin M, Sueda T, Vacanti JP. (2005). Journal of thoracic and cardiovascular surgery. In vitro tissue engineering of a cardiac graft using a degradable scaffold with an extracellular matrix-like topography. 130: 1358 – 1363 Ito A, Hayashida M, Honda H, Hata K-I, Kagami H, Ueda M, Kobayashi T. (2004). Tissue engineering. Construction and harvest of multilayered keratinocyte sheets using magnetite nanoparticles and magnetic force. 10 (5/6): 873 – 880 Ito A, Jitsunobu H, Kawabe Y, Kamihira M. (2007). Journal of bioscience and bioengineering. Construction of heterotypic cell sheets by magnetic force-based 3D coculture of HepG2 and NIH3T3 cells. 104 (5): 371 – 378 Izumi Y, Hirose T, Tamai Y, Hirai S, Nagashima Y, Fujimoto T, Tabuse Y, Kemphues KJ, Ohno S. (1998). Journal of cell biology. An atypical PKC directly associates and colocalizes at the epithelial tight junction with ASIP, a mammalian homologue of Caenorhabditis elegans polarity protein PAR-3. 143 (1): 95 – 106 Jaiswal N, Haynesworth SE, Caplan AI, Bruder SP. (1997). Journal of cellular biochemistry. Osteogenic differentiation of purified, culture-expanded human mesenchymal stem cells in vitro. 64: 295 – 312 Jasmund I, Schwientek S, Acikgoz A, Langsch A, Machens HG, Bader A. (2007). Biomolecular engineering. The influence of medium composition and matrix on longterm cultivation of primary porcine and human hepatocytes. 24 (1): 59 – 69 Karageorgiou V, Kaplan D. (2005). Biomaterials. Porosity of 3D biomaterial scaffolds and osteogenesis. 26 (27): 5475 – 5491 Keating A. (2006). Current opinion in hematology. Mesenchymal stromal cells. 13: 419 – 425 124 Ki CS, Baek DH, Gang KD, Lee KH, Um IC, Park YH. (2005). Polymer. Characterization of gelatin nanofiber prepared from gelatin-formic acid solution. 46: 5094 – 5102 Kim D-H, Kim P, Song I, Cha JM, Lee SH, Kim BK, Suh KY. (2006). Langmuir. Guided three-dimensional growth of functional cardiomyocytes on polyethylene glycol nanostructures. 22: 5419 – 5426 Kim HW, Kim HE, Knowles JC. (2006). Advanced functional materials. Production and potential of bioactive glass nanofibers as a next-generation biomaterial. 16: 1529 – 1535 Kisiday JD, Kurz B, DiMicco MA, Grodzinsky AJ. (2005). Tissue engineering. Evaluation of medium supplemented with insulin-transferrin-selenium for culture of primary bovine calf chondrocytes in three-dimensional hydrogel scaffolds. 11 (1/2): 141 – 151 Ko JA, Liu Y, Yanai R, Chikama T, Takezawa T, Nishida T. (2008). Investigative ophthalmology & visual science. Upregulation of tight-junctional proteins in corneal epithelial cells by corneal fibroblasts in collagen vitrigel cultures. 49 (1): 113 – 119 Koga T, Higuchi M, Kinoshita T, Higashi N. (2006). Chemistry: a European journal. Controlled self-assembly of amphiphilic oligopeptides into shape-specific nanoarchitectures. 12: 1360 – 1367 Kotwal A, Schmidt CE. (2001). Biomaterials. Electrical stimulation alters protein adsorption and nerve cell interactions with electrically conducting biomaterials. 22: 1055 - 1064 Kushida A, Yamato M, Kikuchi A, Okano T. (2001). Journal of biomedical materials research. Two-dimensional manipulation of differentiated Madin-Darby canine kidney (MDCK) cell sheets: the noninvasive harvest from temperature-responsive culture dishes and transfer to other surfaces. 54: 37 – 46 Kwon OH, Nho YC. (2002). Journal of applied polymer science. Radiation-induced graft polymerization of methyl methacrylate onto ultrahigh molecular weight polyethylene in the presence of a metallic salt and acid. 86: 2348 – 2356 Lee SD, Hsiue GH, Chang CT, Kao CY. (1996). Biomaterials. Plasma-induced grafted polymerisation of acrylic acid and subsequent grafting of collagen onto polymer film as biomaterials. 17: 1599 125 Lee YM, Shim JK. (1996). Journal of applied polymer science. Plasma surface graft of acrylic acid onto a porous poly (vinylidene fluoride) membrane and its riboflavin permeation. 61: 1245 Leon EJ, Verma N, Zhang SG, Lauffenburger DA, Kamm RD. (1998). Journal of biomaterial science, polymer edition. Mechanical properties of a self-assembling oligopeptide matrix. (3): 297 – 312 Li D, Xia YN. (2004). Advanced materials. Electrospinning of nanofibers: reinventing the wheel. 16 (14): 1151 – 1170 Li JX, He AH, Han CC, Fang DF, Hsiao BS, Chu B. (2006). Macromolecular rapid communications. Electrospinning of hyaluronic acid (HA) and HA / gelatin blends. 27: 114 – 120 Li MY, Guo Y, Wei Y, MacDiarmid AG, Lelkes PI. (2006). Biomaterials. Electrospinning polyaniline-contained gelatin nanofibers for tissue engineering applications. 27: 2705 – 2715 Lian JB. (2006). Current opinion in endocrinology & diabetes. Biology of bone mineralization. 13: – Liao IC, Wan ACA, Yim EKF, Leong KW. (2005). Journal of controlled release. Controlled release from fibers of polyelectrolyte complexes. 104: 347 – 358 Lim SH, Liao IC, Leong KW. (2006). Molecular therapy. Nonviral gene delivery from nonwoven fibrous scaffolds fabricated by interfacial complexation of polyelectrolytes. 13 (6): 1163 – 1172 Lou YR, Miettinen S, Kagechika H, Gronemeyer H, Tuohimaa P. (2005). Biochemical and biophysical research communications. Retinoic acid via RARα inhibits the expression of 24-hydroxylase in human prostate stromal cells. 338: 1973 – 1981 Ma PX, Zhang RY. (1999). Journal of biomedical materials research. Synthetic nanoscale fibrous extracellular matrix. 46: 60 – 72 Ma ZW, Gao CY, Shen JC. (2003). Journal of biomaterial science: polymer edition. 126 Surface modification of poly-L-lactic acid (PLLA) membrane by grafting acrylamide: an effective way to improve cytocompatibility for chondrocytes. 14 (1): 13 - 25 Ma ZW, Kotaki M, Yong T, He W, Ramakrishna S. (2005). Biomaterials. Surface engineering of electrospun polyethylene terephthalate (PET) nanofibers towards development of a new material for blood vessel engineering. 26: 2527 – 2536 Maher JJ, Bissell DM. (1993). Seminars in cell biology. Cell-matrix interactions in liver. 4: 189 – 201 Margadant C, Opstal AV, Boonstra J. (2007). Journal of cell science. Focal adhesion signaling and actin stress fibers are dispensable for progression through the ongoing cell cycle. 120: 66 – 76 McCook A. (2007). The scientist. Betting on better organs. 21(12): 31 – 35 McKee MG, Wilkes GL, Colby RH, Long TE. (2004). Macromolecules. Correlations of solution rheology with electrospun fiber formation of linear and branched polyesters. 37: 1760 – 1767 McManus MC, Boland ED, Simpson DG, Barnes CP, Bowlin GL. (2007). Journal of biomedical materials research. Electrospun fibrinogen: feasibility as a tissue engineering scaffold in a rat cell culture model. 81A: 299 – 309 Mit-uppatham C, Nithitanakul M, Supaphol P. (2004). Macromolecular chemistry and physics. Ultrafine electrospun polyamide-6-fibers: effect of solution conditions on morphology and average fiber diameter. 205: 2327 – 2338 Moreno-Borchart A. (2004). EMBO reports. Building organs piece by piece. 5(11): 1025 – 1028. Murugan R, Ramakrishna S. (2006). Tissue engineering. Nano-featured scaffolds for tissue engineering: a review of spinning methodologies. 12 (3): 435 – 447 Murugan R, Ramakrishna S. (2007). Tissue engineering. Design strategies of tissue engineering scaffolds with controlled fiber orientation. 13 (8): 1845 – 1866 127 Nagai Y, Unsworth LD, Koutsopoulos S, Zhang SG. (2006). Journal of controlled release. Slow release of molecules in self-assembling peptide nanofiber scaffold. 115: 18 – 25 Narkis M, Sibony-Chaouat S, Siegmann A, Shkolnik S, Bell JP. (1985). Polymer. Irradiation effects on polycaprolactone. 26: 50 Nature biotechnology review. Tissue engineering. 18 supplement 2000: IT56 – 58. Nerem RM. (2006). Tissue engineering. Tissue engineering: the hope, the hype and the future. 12 (5): 1143 – 1150 Ng CS, Teoh SH, Chung TS, Hutmacher DW. (2000). Polymer. Simultaneous biaxial drawing of poly (ε-caprolactone) films. 41: 5855 – 5864 Ng S, Han RB, Chang S, Ni J, Hunziker W, Goryachev AB, Ong SH, Yu H. (2006). Tissue engineering. Improved hepatocyte excretory function by immediate presentation of polarity cues. 12(8): 2181 – 2191 Ohashi K, Yokoyama T, Yamato M, Kuge H, Kanehiro H, Tsutsumi M, Amanuma T, Iwata H, Yang J, Okano T, Nakajima Y. (2007). Nature medicine. Engineering functional two- and three-dimensional liver systems in vivo using hepatic tissue sheets. 13 (7): 880 – 885 Oshio C, Phillips MJ. (1981). Science. Contractility of bile canaliculi: implications for liver function. 212 (4498): 1041 – 1042 Palmer J. (2000). Barron. Spare body parts. Paredes R, Arriagada G, Cruzat F, Olate J, Wijnen AV, Lian J, Stein G, Stein J, Montecino M. (2004). Journal of steroid biochemistry & molecular biology. The Runx2 transcription factor plays a key role in the 1α, 25-dihydroxy vitamin D3dependent upregulation of the rat osteocalcin (OC) gene expression in osteoblastic cells. 89 – 90: 269 – 271 Pece S, Gutkind JS. (2000). Journal of biological chemistry. Signaling from Ecadherins to the MAPK pathway by the recruitment and activation of epidermal growth factor receptors upon cell-cell contact formation. 275 (52): 41227 – 41233 128 Pham QP, Sharma U, Mikos AG. (2006). Biomacromolecules. Electrospun poly (εcaprolactone) microfiber and multilayer nanofiber / microfiber scaffolds: characterization of scaffolds and measurement of cellular infiltration. 7: 2796 – 2805 Rajagopalan P, Shen CJ, Berthiaume F, Tilles AW, Toner M, Yarmush ML. (2006). Tissue engineering. Polyelectrolyte nano-scaffolds for the design of layered cellular architectures. 12 (6): -11 Rosenberg MD. (1963). Science. Cell guidance by alterations in monomolecular films. 139: 411 Sarikaya M, Tamerler C, Jen AKY, Schulten K, Baneyx F. (2003). Nature materials. Molecular biomimetics: nanotechnology through biology. 2: 577 – 585 Schwartz RE, Reyes M, Koodie L, Jiang YH, Blackstad M, Lund T, Lenvik T, Johnson S, Hu W-S, Verfaillie CM. (2002). Journal of clinical investigation. Multipotent adult progenitor cells from bone marrow differentiate into functional hepatocyte-like cells. 109: 1291 - 1302 Sekiya I, Larson BL, Smith JR, Pochampally R, Cui JG, Prockop DJ. (2002). Stem cells. Expansion of human adult stem cells from bone marrow stroma: conditions that maximize the yields of early progenitors and evaluate their quality. 20: 530 – 541 Selden C, Hodgson H. (2004). Transplant immunology. Cellular therapies for liver replacement. 12 (3 – 4): 273 – 288 Sell S, Barnes C, Simpson D, Bowlin G. (2008). Journal of biomedical materials research A. Scaffold permeability as a means to determined fiber diameter and pore size of electrospun fibrinogen. 85A (1): 115 – 126 Semino CE, Merok JR, Crane GG, Panagiotakos G, Zhang SG. (2003). Differentiation. Functional differentiation of hepatocyte-like spheroid structures from putative liver progenitor cells in three-dimensional peptide scaffolds. 71: 262 – 270 Sexton LT, Horne LP, Martin CR. (2007). Molecular biosystems. Developing synthetic conical nanopores for biosensing applications. 3: 667 – 685 Shastri VP. (2006). Artificial organs. Future of regenerative medicine: challenges and hurdles. 30(10): 828 – 834 129 Shenoy SL, Bate WD, Frisch HL, Wnek GE. (2005). Polymer. Role of chain entanglements on fiber formation during electrospinning of polymer solutions: good solvent, non-specific polymer-polymer interaction limit. 46: 3372 – 3384 Shi YC, Worton L, Esteban L, Baldock P, Fong C, Eisman JA, Gardiner EM. (2007). Bone. Effects of continuous activation of vitamin D and Wnt response pathways on osteoblastic proliferation and differentiation. 41 (1): 87 – 96 Shin M, Ishii O, Sueda T, Vacanti JP. (2004). Biomaterials. Contractile cardiac grafts using a novel nanofibrous mesh. 25: 3717 – 3723 Shin YM, Hohman MM, Brenner MP, Rutledge GC. (2001). Applied physics letters. Electrospinning: a whipping fluid jet generates submicron polymer fibers. 78 (8): 1149 - 1151 Sierra J, Villagra A, Paredes R, Cruzat F, Gutierrez S, Javed A, Arriagada G, Olate J, Imschenetzky M, Van Wijnen AJ, Lian JB, Stein GS, Stein JL, Montecino M. (2003). Molecular and cellular biology. Regulation of the bone-specific osteocalcin gene by p300 requires Runx2 / Cbfa1 and the vitamin D3 receptor but not p300 intrinsic histone acetyltransferase activity. 23 (9): 3339 – 3351 Silva GA, Czeisler C, Niece KL, Beniash E, Harrington DA, Kessler JA, Stupp SL. (2004). Science. Selective differentiation of neural progenitor cells by high-epitope density nanofibers. 303: 1352 – 1355 Sniadecki NJ, Desai RA, Ruiz SA, Chen CS. (2006). Annals of biomedical engineering. Nanotechnology for cell-substrate interactions. 34 (1): 59 – 74 Sodergard A. (1998). Journal of polymer science: polymer chemistry. Preparation of poly(ε-caprolactone)-co-poly(acrylic acid) by radiation-induced grafting. (36): 1805 Stains JP, Lecanda F, Screen J, Towler DA, Civitelli R. (2003). Journal of biological chemistry. Gap junctional communication modulates gene transcription by altering the recruitment of Sp1 and Sp3 to connexin-response elements in osteoblast promoters. 278 (27): 24377 – 24387 Stankus JJ, Guan JJ, Fujimoto K, Wagner WR. (2006). Biomaterials. Microintegrating smooth muscle cells into a biodegradable, elastomeric fiber matrix. 27: 735 – 744 130 Stankus JJ, Soletti L, Fujimoto K, Hong Y, Vorp DA, Wagner WR. (2007). Biomaterials. Fabrication of cell microintegrated blood vessel constructs through electrohydrodynamic atomization. 28: 2738 – 2746 Stevens MM, George JH. (2005). Science. Exploring and engineering the cell surface interface. 310: 1135 – 1138 Stock UA, Vacanti JP. (2001). Annual review in medicine. Tissue engineering: current state and prospects. 52: 443 – 451 Sun DH, Chang C, Li S, Lin LW. (2006). Nano letters. Near-field electrospinning. (4): 839 – 842 Takezawa T, Ozaki K, Nitani A, Takabayashi C, Shimo-Oka T. (2004). Cell transplantation. Collagen vitrigel: a novel scaffold that can facilitate a threedimensional culture for reconstructing organoids. 13: 463 – 473 Takezawa T, Ozaki K, Takabayashi C. (2007). Tissue engineering. Reconstruction of a hard connective tissue utilizing a pressed silk sheet and type-I collagen as the scaffold for fibroblasts. 13 (6): 1357 – 1366 Takezawa T, Takeuchi T, Nitani A, Takayama Y, Kino-oka M, Taya M, Enosawa S. (2007). Journal of Biotechnology. Collagen vitrigel membrane useful for paracrine assays in vitro and drug delivery systems in vivo. 131: 76 – 83 Teo WE, Kotaki M, Mo XM, Ramakrishna S. (2005). Nanotechnology. Porous tubular structures with controlled fibre orientation using a modified electrospinning method. 16: 918 – 924 Thompson CJ, Chase GG, Yarin AL, Reneker DH. (2007). Polymer. Effects of parameters on nanofiber diameter determined from electrospinning model. 48: 6913 – 6922 Tiaw KS, Goh SW, Hong M, Wang Z, Lan B, Teoh SH. (2005). Biomaterials. Laser surface modification of poly (ε-caprolactone) (PCL) membrane for tissue engineering applications. 26: 763 – 769 Tiaw KS, Teoh SH, Chen R, Hong MH. (2007). Biomacromolecules. Processing methods of ultrathin poly(ε-caprolactone) films for tissue engineering applications. 8: 807 – 816 131 Toh YC, Ng S, Khong YM, Zhang X, Zhu YJ, Lin PC, Te CM, Sun WX, Yu H. (2006). Nanotoday. Cellular responses to a nanofibrous environment. (3): 34 – 43 Tsukada N, Ackerley CA, Phillips MJ. (1995). Hepatology. The structure and organization of the bile canalicular cytoskeleton with special reference to actin and actin-binding proteins. 21 (4): 1106 – 1113 Tuli R, Tuli S, Nandi S, Huang XX, Manner PA, Hozack WJ, Danielson KG, Hall DJ, Tuan RS. (2003). Journal of biological chemistry. Transforming growth factor-βmediated chondrogenesis of human mesenchymal progenitor cells involves Ncadherin and mitogen-activated protein kinase and Wnt signaling cross-talk. 278 (42): 41227 – 41236 Vacanti CA. (2006). Tissue engineering. History of tissue engineering and a glimpse into its future. 12 (5): 1137 – 1142 Van Der Heyden MAG, Rook MB, Hermans MMP, Rijksen G, Boonstra J, Defize LHK, Destree OHJ. (1998). Journal of cell science. Identification of connexin43 as a functional target for Wnt signaling. 111: 1741 – 1749 Vauthey S, Santoso S, Gong HY, Watson N, Zhang SG. (2002). Proceedings of the National Academy of Sciences of the United States of America. Molecular selfassembly of surfactant-like peptides to form nanotubes and nanovesicles. 99 (8): 5355 – 5360 Vinken M, Papeleu P, Snykers S, De Rop E, Henkens T, Chipman JK, Rogiers V, Vanhaecke T. (2006). Critical reviews in toxicology. Involvement of cell junctions in hepatocyte culture functionality. 36 (4): 299 – 318. Wan ACA, Liao IC, Yim EKF, Leong KW. (2004). Macromolecules. Mechanism of fiber formation by interfacial polyelectrolyte complexation. 37: 7019 – 7025 Wan ACA, Tai BCU, Leck KJ, Ying JY. (2006). Advanced materials. Silicaincorporated polyelectrolyte-complex fibers as tissue-engineering scaffolds. 18: 641 – 644 Wan ACA, Yim EKF, Liao IC, Visage CL, Leong KW. (2004). Journal of biomedical materials research. Encapsulation of biologics in self-assembled fibers as biostructural units for tissue engineering. 71A: 586 – 595 132 Wang LG, Topham PD, Mykhaylyk OO, Howse JR, Bras W, Jones RAL, Ryan AJ. (2007). Advanced materials. Electrospinning pH-responsive block copolymer nanofibers. 19: 3544 – 3548 Wang YJ, Liu HL, Guo HT, Wen HW, Liu J. (2004). World journal of gastroenterology. Primary hepatocyte culture in collagen gel mixture and collagen sandwich. 10(5): 699 - 702 Weitz RT, Harnau L, Rauschenbach S, Burghard M, Kern K. (2008). Nano letters. Polymer nanofibers via nozzle-free centrifugal spinning. (4): 1187 – 1191. Wilkinson RC, Dickson AJ. (2001). Biochemical and biophysical research communications. Expression of CCAAT / enhancer binding protein family genes in monolayer and sandwich culture of hepatocytes: induction of stress-inducible GADD153. 289: 942 – 949 Woo KM, Chen VJ, Ma PX. (2003). Journal of biomedical materials research. Nanofibrous scaffolding architecture selectively enhances protein adsorption contributing to cell attachment. 67A: 531 – 537 Woodfield TB, Van Blitterswijk CA, De Wijn J, Sims TJ, Hollander AP, Riesle J. (2005). Tissue engineering. Polymer scaffolds fabricated with pore-size gradients as a model for studying the zonal organization within tissue-engineered cartilage constructs. 11 (9-10): 1297 – 1311 Xia W, Zhang DM, Chang J. (2007). Nanotechnology. Fabrication and in vitro biomineralization of bioactive glass (BG) nanofibers. 18: – Xu CY, Inai R, Kotaki M, Ramakrishna S. (2004). Biomaterials. Aligned biodegradable nanofibrous structure: a potential scaffold for blood vessel engineering. 25: 877 – 886 Yamamoto H, Horita C, Senoo Y, Nishida A, Ohkawa K. (2001). Journal of applied polymer science. Polyion complex fiber and capsule formed by self-assembly of polyl-lysine and gellan at solution interfaces. 79: 437 – 446 Yang J, Yamato M, Nishida K, Ohki T, Kanzaki M, Sekine H, Shimizu T, Okano T. (2006). Journal of controlled release. Cell delivery in regenerative medicine: the cell sheet engineering approach. 116: 193 – 203 133 Ye Q, Zund G, Benedikt P, Jockenhoevel S, Hoerstrup SP, Sakyama S, Hubbell JA, Turina M. (2000). European journal of cardio-thoracic surgery. Fibrin gel as a three dimensional matrix in cardiovascular tissue engineering. 17: 587 – 591 Yeaman C, Grindstaff KK, Nelson WJ. (1999). Physiological reviews. New perspectives on mechanisms involved in generating epithelial cell polarity. 79 (1): 73 – 98 Yeong WY, Chua CK, Leong KF, Chandrasekaran M. (2004). Trends in biotechnology. Rapid prototyping in tissue engineering: challenges and potential. 22 (12): 643 – 652 Yim EKF, Wan ACA, Visage CL, Liao IC, Leong KW. (2006). Biomaterials. Proliferation and differentiation of human mesenchymal stem cell encapsulated in polyelectrolyte complexation fibrous scaffold. 27: 6111 – 6122 Yu HM, Meyvantsson I, Shkel IA, Beebe DJ. (2005). Lab on a chip. Diffusion dependent cell behavior in microenvironments. 5: 1089 – 1095 Yu JH, Fridrikh SV, Rutledge GC. (2006). Polymer. The role of elasticity in the formation of electrospun fibers. 47: 4789 – 4797 Zhang H, Hutmacher DW, Chollet F, Poo AN, Burdet E. (2005). Macromolecular bioscience. Microrobotics and MEMS-based fabrication techniques for scaffold-based tissue engineering. 5: 477 – 489 Zhang K, Wang Y, Hillmyer MA, Francis LF. (2004). Biomaterials. Processing and properties of porous poly (l-lactide) / bioactive glass composites. 25 (13): 2489 – 2500. Zhang RY, Ma PX. (2000). Journal of biomedical materials research. Synthetic nanofibrillar extracellular matrices with predesigned macroporous architectures. 52: 430 – 438 Zhang SG, Marini DM, Hwang WM, Santoso S. (2002). Current opinion in chemical biology. Design of nanostructured biological materials through self-assembly of peptides and proteins. 6: 865 – 871 134 Zhang SG. (2002). Biotechnology advances. Emerging biological materials through molecular self-assembly. 20: 321 – 339 Zhang SG. (2003). Nature biotechnology. Fabrication of novel biomaterials through molecular self-assembly. 21 (10): 1171 – 1178 Zhang Y, Chai C, Jiang XS, Teoh SH, Leong KW. (2007). Materials science & engineering c – biomimetic and supramolecular systems. Fibronectin immobilized by covalent conjugation or physical adsorption shows different bioactivity on aminatedPET. 27 (2): 213 – 219 Zhao XJ, Zhang SG. (2004). Trends in biotechnology. Fabrication of molecular materials using peptide construction motifs. 22 (9): 470 – 476 Zhou YF, Sae-Lim V, Chou AM, Hutmacher DW, Lim TM. (2006). Journal of biomedical materials research. Does seeding density affect in vitro mineral nodules formation in novel composite scaffolds. 78A: 183 - 193 Zhu JH, Wang XW, Ng S, Quek CH, Ho HT, Lao XJ, Yu H. (2005). Journal of biotechnology. Encapsulating live cells with water soluble chitosan in physiological conditions. 117: 355 – 365 Zipori D. (2004). Blood cells, molecules and diseases. Mesenchymal stem cells: harnessing cell plasticity to tissue and organ repair. 33: 211 – 215 135 [...]... domains that allow them to self assemble onto a surface instead of among themselves The first component is usually a terminal segment of functional groups that other bioactive agents recognize; the second is a flexible spacer (usually a string of hydrophobic amino acids such as alanine or valine) to minimize non-specific binding; the third is a cysteine, asparatic acid or lysine residue that can covalently... addition, degradation kinetics of these phaseseparated scaffolds was also found to be significantly faster in spite of the hydrophobicity as a result of more sites being made available for hydrolysis due to a 100-fold increase in surface area (Chen VJ, 2006) In fact, loss in mass for the first 12 months was determined to be as high as 50 % Parallel fabrication of a multitude of nanofibers with regular morphology... proteases Insulin-like growth factor-1 tethered to the peptide scaffold via a biotin / streptavidin sandwich approach remained capable of inducing in vivo Akt phosphorylation and cardiomyocytes maturation even after two weeks (Davis ME, 2006) 2.2.4 Electrospinning Among all the techniques available, electrospinning offers one of the simplest and cost-effective ways to manufacture nano-sized features rapidly... mineralizations whose crystallographic c axes were aligned along the long axes of fibers like those at native bone tissues (Hartgerink JD, 2001) Apart from facilitating crystal growth via the incorporation of phosphoserine and aspartic acid, these nucleations were also initiated by concentrating the inorganic cations to local supersaturation on surfaces where repeating anionic groups were exposed Interfacing... from the host (probably due to their ionic nature), they were also found to be biodegradable in an in- vivo environment despite their in- vitro stability towards heat, chemicals and proteolytic enzymes (Leon EJ, 1998) SAPs are also amenable to further modification via a level as small as one amino acid Biologically active domains can therefore be left flagging via a glycine spacer on the C-termini of one... manipulating the physico-chemical properties from a library of 20 natural amino acids and a growing number of artificial derivatives (Leon EJ, 1998) Three theoretical models are currently in use to enable better understanding of the assembly process (Zhang SG et al, 2002) The first makes use of molecular dynamics simulation to explore the packing order between the peptides and the energy landscape associated... multidisciplinary field also known as TE where various aspects of medicine, materials science, engineering and biology are combined to help persuade the body to regenerate itself through the use of cells, supporting structure and biomolecules Ironically, its humble beginning was traced back not to any scientific journals, but rather a famous “Healing of Justinian” 18 portrait by Italian renaissance painter Fra... channels for infiltration to take place either during seeding or in vivo tissue remodeling events such as neural / vasculature anastomosis The scaffold should also exhibit appropriate mechanical properties: a substrate too rigid will result in the inability of the cells to recognize and recruit the receptors into focal adhesion plaques for signal propagations; on the other hand, a material too compliant will... electrical double-layer repulsion (Caplan MR, 2002) The latter was determined to constitute a kinetic barrier that must be minimized to near kT by charge screening from the salts in order to allow the approach of the oligopeptides Not only were the resulting hydrogels highly hydrated, structural characterizations also revealed matching porosity with their native analogs In addition to the lack of severe... barriers around electrodes and scarring of heart valve sewing rings are just some of the activities that had reportedly led to implant failure As a result, not even durable items like hip or knee prostheses are able to last beyond 15 years There are also methods such as radical cystectomy that attempt to substitute the malfunctioning organ (bladder) with a similar analog (intestinal tissue) in the host . rather a famous “Healing of Justinian” 19 portrait by Italian renaissance painter Fra Angelica (1400 – 1455) where two brothers Saints Damien and Cosmos were drawn in the process of transplanting. permeable material with ease of handling can therefore be fabricated and its potential use as a sandwiching or stacking membrane was examined. Morphological and functionality studies on overlaid. mechanical wear or secondary inflammatory responses (Hutmacher DW, 2006). Capsular contracture of breast implants, insulating barriers around electrodes and scarring of heart valve sewing rings are

Ngày đăng: 11/09/2015, 16:04

TỪ KHÓA LIÊN QUAN

TÀI LIỆU CÙNG NGƯỜI DÙNG

TÀI LIỆU LIÊN QUAN

w