帳號:guest(3.17.181.21)          離開系統
字體大小: 字級放大   字級縮小   預設字形  

詳目顯示

以作者查詢圖書館館藏以作者查詢臺灣博碩士論文系統以作者查詢全國書目
作者(中文):葉怡君
作者(外文):Yeh, Yi-Chun
論文名稱(中文):可注射式細胞載體於心肌組織工程的應用
論文名稱(外文):Injectable Cell Delivery Vehicles for Myocardial Tissue Engineering
指導教授(中文):宋信文
指導教授(外文):Sung, Hsing-Wen
學位類別:博士
校院名稱:國立清華大學
系所名稱:化學工程學系
學號:9532826
出版年(民國):99
畢業學年度:99
語文別:英文
論文頁數:91
中文關鍵詞:細胞治療羊水幹細胞細胞片工程可注射式載體
相關次數:
  • 推薦推薦:0
  • 點閱點閱:110
  • 評分評分:*****
  • 下載下載:2
  • 收藏收藏:0
Myocardial infarction (MI) progresses from the acute death of cardiomyocytes and the infiltration of inflammatory cells into granulation, followed by scars. Recently, cell transplantation via local intramuscular injection is a promising therapy for patients with MI. However, the time required to process and expand autologous cells in vitro makes applying these cells in patients with an acute MI in a timely fashion difficult. Moreover, following intramuscular injection, retention of the dissociated cells in the transplanted area remains problematic. These facts can be deleterious to cell transplantation therapy.
The purpose of study I to evaluate using human amniotic fluid stem cells (hAFSCs) derived from second-trimester amniocentesis for the therapeutic potential of cardiac repair. Whether hAFSCs can be differentiated into cardiomyogenic cells and toward the maturation of endothelial cell lineage was investigated in vitro using mimicking differentiation milieu. Test samples were xenogenically transplanted into the peri-ischemia area of an immune-suppressed rat model at 1 week after MI induction. There were three treatment groups (n ≥ 10): sham; saline; dissociated hAFSCs. After 4 weeks, hAFSCs-treated animals showed a preservation of the infracted thickness, an attenuation of left ventricle remodeling, a higher vascular density, and thus an improvement in cardiac function.
Study II examined the hypothesis that the thermoreversible methylcellulose hydrogel was used as a coating on TCPS dishes and developed for harvesting living hAFSCs sheets. The optioned hAFSCs sheets preserved the intercellular junctions and endogenous extracellular matrix (ECM) and retained the cell phenotype. Test samples were xenogenically transplanted into the peri-ischemia area of an immune-suppressed rat model at 1 week after MI induction. There were four treatment groups (n ≥ 10): sham; saline; dissociated hAFSCs; and hAFSCs sheet fragments. The results obtained in the echocardiography revealed that the group treated with hAFSCs-sheet-fragment had a superior heart function to those treated with saline and dissociated hAFSCs group (P < 0.05). Due to their inherent ECM, hAFSCs sheet fragments had a better ability of cell retention and proliferation than dissociated hAFSCs upon transplantation to the host myocardium. Additionally, transplantation of the hAFSCs sheet fragments stimulated a signification increase in vascular density, consequently contributing towards improved wall thickness and a reduction in the infarct size. Our histological findings and qPCR analyses suggest that the transplanted hAFSCs can be differentiated into cardiomyocyte-like cells and cell of endothelial lineaged and modulate expression of multiple angiogenic cytokines and cardiac protective factor with the potential to promote neo-vascularization, which evidently contributed to the improvement of ventricular function. These results suggest that the use of fragments of cell sheets can serve as a cell-delivery vehicle by providing an adequate physical size and a favorable ECM environment to retain the transplanted hAFSCs in the infracted myocardium. The benefit effects of hAFSCs may be mediated not only through their ability to differentiate into cardiomyocyte-like cells and cells of endothelial lineages but also through the growth factor-mediated paracrine regulation.
In our previous study, the number of cells on the 50-mesh-sized for 27 gauge optimize injection condition is 65 cells/fragment. The ideal number of cell transplantation for clinical use is around from 1×106 to 1×107. Recently, the use of three dimensional multicellular aggregates (cell bodies) for myocardium infarction was investigated. For the human amniotic fluid stem cells, 1×104 cells on the methylcellulose hydrogel can form the cell body and the diameter can reach to 350 μm. However, it has been often reported that cell necrosis occurs due to the insufficient supply of oxygen/nutrients and the accumulation of metabolic wastes, especially at the center of the cell bodies. In order to address this problem, the injectable of three dimensional porous micro-scaffolds were developed to enhance the transportation of oxygen/nutrients and wastes. In the study III, uniform porous poly(D, L-lactide-co-glycolide) (PLGA) beads with controlled pore diameters were prepared based on an emulsion templating method in a simple fluidic device. Unstable water-in-oil (W-O) emulsion, being in the course of phase-separation, was introduced into the fluidic device to form water-in-oil-in-water (W-O-W) droplets. The resultant W-O-W droplets evolved into porous PLGA beads with controlled pore diameters at the inside and surface of the beads after the organic solvent had evaporated. Porous beads prepared using the upper part of the phase-separated emulsion exhibited the large inner and surface pores as well as high interconnectivity compared to the lower part of the emulsions. Fluorescence microscopy imaging and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay confirmed the more uniform distribution and higher viability of the cells in the bead with large pores. These results suggest that the bead with large pores could provide favorable environment for cells and thus be potentially used for both tissue engineering and cell delivery system.
ABSTRACT I
TABLE OF CONTENT III
LIST OF FIGURES AND TABLES VII

Chapter 1. Introduction 1

Chapter 2.
Cellular Cardiomyoplasty with Human Amniotic FluidStem Cells: In Vitro and In Vivo Studies
2-1 Introduction ----------6
2-2 Materials and Methods 7
2-2.1. Characterization of hAFSC 7
2-2.2. Differentiation of hAFSC into endothelial-like cells in vitro-8
2-2.3. Cardiomyogenic differentiation of hAFSC in vitro 8
2-2.4. Animal study 11
2-2.5. LV function assessment by echocardiography 12
2-2.6. Histological examinations 13
2-2.7. Statistical analyses 15
2-3 Results 15
2-3.1. Charscterization of hAFSC 15
2-3.2. Differentiation of hAFSC into endothelial-like cells in vitro
--------------------------------------------------------------------------------16
2-3.3. Cardiomyogenic differentiation of hAFSC in vitro 17
2-3.4. Animal study 20
2-3.5.Cardiac function 20
2-3.6.Morphological observations 23
2-3.7.Histological findings 24
2-4. Discussion -----------------------------------------------------------------28
2-5. Conclusions 32

Chapter 3.
Cardiac Repair with Injectable Cell Sheet Fragments of Human Amniotic Fluid Stem Cells in an Immune-Suppressed Rat Model
3-1. Introduciton ----------------------------------------------------------------33
3-2. Materials and Methods 36
3-2.1. Characterization of isolated hAFSCs--------------------------36
3-2.2. Construction and characterization of hAFSC sheet fragments
-------------------------------------------------------------------------------37
3-2.3. Cell transplantations----------------------------------------------38
3-2.4. Left ventricular function assessment by echocardiography
------------------------------------------------------------------------------39
3-2.5. Histological examinations---------------------------------------40
3-2.6. Quantitative real-time polymerase chain reaction (qPCR) analyses------------------------------------------------------------------------42
3-2.7. Statistical analysis 42
3-3 Results and Discussion 42
3-3.1. Characterization of isolated hAFSCs 43
3-3.2. Characteristics of hAFSCs sheet fragments 43
3-3.3. Animal study 46
3-3.4. LV function 46
3-3.5. Morphological observations 48
3-3.6. Histological findings 50
3-3.7. qPCR analyses 55
3-4 Conclusions 58

Chapter 4.
Uniform PLGA Beads with Controllable Pore Sizes for Biomedical Applications
4-1 Introduction 59
4-2 Materials and Methods 61
4-2.1. Materials 61
4-2.2. Preparation and characterization of porous beads using the fluidic device 62
4-2.3 In Vitro cell culture study 63
4-2.4 Cell viability 64
4-3 Results and Discussion 65
4-3.1 Fabrication of porous beads using fluidic device 65
4-3.2 Characterization of porous beads using fluidic device 68
4-3.3 Cell culture with porous beads of different pore sizes 72
4-4 Conclusions 78

References 79
[1] Ryu JH, Kim IK, Cho SW, Cho MC, Hwang KK, Piao H, et al. Implantation of bone marrow mononuclear cells using injectable fibrin matrix enhances neovascularization in infarcted myocardium. Biomaterials 2005; 26:319-26
[2] Perin EC, Dohmann HF, Borojevic R, Silva SA, Sousa AL, Mesquita CT, et al. Transendocardial, autologous bone marrow cell transplantation for severe, chronic ischemic heart failure. Circulation 2003;107:2294-302.
[3] Kim WG, Park JJ, Chung DH, and Na CY. Autologous cardiomyocyte transplantation in an ovine myocardial infarction model. Int J Artif Organs 2002;25:61-6.
[4] Smits AM, van Laake LW, den Ouden K, Schreurs C, Szuhai K, van Echteld CJ, et al. Human cardiomyocyte progenitor cell transplantation preserves long-term function of the infarcted mouse myocardium. Cardiovasc Res 2009;83:527-35.
[5] Yoo, KJ, Kim, HO, Kwak, YL, Kang, SM, Jang YS, Lim SH, et al. Autologous bone marrow cell transplantation combined with off-pump coronary artery bypass grafting in patients with ischemic cardiomyopathy. Can J Surg 2008;51:269-75.
[6] Zhao P, Ise H, Hongo M, Ota M, Konishi I, and Nikaido T. Human amniotic mesenchymal cells have some characteristics of cardiomyocytes. Transplantation 2005;79:528-35.
[7] Chiavegato A, Bollini S, Pozzobon M, Callegari A, Gasparotto L, Taiani J, et al. Human amniotic fluid-derived stem cells are rejected after transplantation in the myocardium of normal, ischemic, immuno-suppressed or immuno-deficient rat. J Mol Cell Cardiol 2007;42:746-59.
[8] Freshney RI. Culture of animal cells: A manual of basic technique. 3rd ed. New York: Wiley-Liss; 1994.
[9] Memon IA, Sawa Y, Fukushima N, Matsumiya G, Miyagawa S, Taketani S, et al. Repair of impaired myocardium by means of implantation of engineered autologous myoblast sheets. J Thorac Cardiovasc Surg 2005;130:1333–41.
[10] Teng CJ, Luo J, Chiu RC, Shum-Tim D. Massive mechanical loss of microspheres with direct intramyocardial injection in the beating heart: Implications for cellular cardiomyoplasty. J Thorac Cardiovasc Surg 2006;132:628–32.
[11] Chen CH, Chang Y, Wang CC, Huang CH, Huang CC, Yeh YC, et al. Construction and characterization of fragmented mesenchymal-stem-cell sheets for intramuscular injection. Biomaterials 2007;28:4634–51.
[12] Tsai MS, Lee JL, Chang YJ, and Hwang SM. Isolation of human multipotent mesenchymal stem cells from second-trimester amniotic fluid using a novel two-stage culture protocol. Hum Reprod 2004;19:1450-6.
[13] Yang J, Yamato M, Kohno C, Nishimoto A, Sekine H, Fukai F, et al. Cell sheet engineering: recreating tissues without biodegradable scaffolds. Biomaterials. 2005;26:6415-22.
[14] Lim SM, Lee HJ, Oh SH, Kim JM, Lee JH. Novel fabrication of PCL porous beads for use as an injectable cell carrier system. J Biomed Mater Res B Appl Biomater. 2009;90:521-30.
[15] Ma PX, Zhang R, Xiao G, Franceschi R. Engineering new bone tissue in vitro on highly porous poly(alpha-hydroxyl acids)/hydroxyapatite composite scaffolds. J Biomed Mater Res. 2001;54:284-93.
[16] Sikavitsas VI, Bancroft GN, Mikos AG. Formation of three-dimensional cell/polymer constructs for bone tissue engineering in a spinner flask and a rotating wall vessel bioreactor. J Biomed Mater Res. 2002;62:136-48.
[17] Sahoo SK, Panda AK, Labhasetwar V. Characterization of porous PLGA/PLA microparticles as a scaffold for three dimensional growth of breast cancer cells. Biomacromolecules 2005;6:1132-9.
[18] Furukawa KS, Suenaga H, Toita K, Numata A, Tanaka J, Ushida T, et al. Rapid and large-scale formation of chondrocyte aggregates by rotational culture. Cell Transplant 2003;12:475-9.
[19] Matsuno T, Hashimoto Y, Adachi S, Omata K, Yoshitaka Y, Ozeki Y, et al. Preparation of injectable 3D-formed beta-tricalcium phosphate bead/alginate composite for bone tissue engineering. Dent Mater J 2008;27:827-34.
[20] Senuma Y, Franceschin S, Hilborn JG, Tissieres P, Bisson I, Frey P. Bioresorbable microspheres by spinning disk atomization as injectable cell carrier: from preparation to in vitro evaluation. Biomaterials 2000;21:1135-44.
[21] Eiselt P, Yeh J, Latvala RK, Shea LD, Mooney DJ, Porous carriers for biomedical applications based on alginate hydrogels. Biomaterials 2000;21:1921-7.
[22] McGlohorn JB, Grimes LW, Webster SS, Burg KJ. Characterization of cellular carriers for use in injectable tissue-engineering composites. J Biomed Mater Res A 2003;66:441-9.
[23] Kim TK, Yoon JJ, Lee DS, Park TG. Gas foamed open porous biodegradable polymeric microspheres. Biomaterials 2006;27:152-9.
[24] Chung HJ, Park TG. Injectable cellular aggregates prepared from biodegradable porous microspheres for adipose tissue engineering. Tissue Eng Part A 2009;15:1391-400.
[25] Chung HJ, Kim IK, Kim TG, Park TG. Highly open porous biodegradable microcarriers: in vitro cultivation of chondrocytes for injectable delivery. Tissue Eng Part A. 2008;14:607-15.
[26] Wolbank S, Stadler G, Peterbauer A, Gillich A, Karbiener M, Streubel B, et al. Telomerase immortalized human amnion- and adipose-derived mesenchymal stem cells: maintenance of differentiation and immunomodulatory characteristics. Tissue Eng Part A 2009;15:1843-54.
[27] De Coppi P, Bartsch G Jr, Siddiqui MM, Xu T, Santos CC, Perin L, et al. Isolation of amniotic stem cell lines with potential for therapy. Nat Biotechnol 2007;25:100-6.
[28] Iop L, Chiavegato A, Callegari A, Bollini S, Piccoli M, Pozzobon M, et al. Different cardiovascular potential of adult- and fetal-type mesenchymal stem cells in a rat model of heart cryoinjury. Cell Transplant 2008;17: 679-94.
[29] Peister A, Deutsch ER, Kolambkar Y, Hutmacher DW, Guldberg RE. Amniotic Fluid Stem Cells Produce Robust Mineral Deposits on Biodegradable Scaffolds. Tissue Eng Part A 2009;15:3129-38.
[30] Zhang P, Baxter J, Vinod K, Tulenko TN, and Dimuzio P. Endothelial Differentiation of Amniotic Fluid-derived Stem Cells: Synergism of biochemical and shear force stimuli. Stem Cells Dev 2009;18:1299-308.
[31] Tsai MS, Hwang SM, Tsai YL, Cheng FC, Lee JL, and Chang YJ. Clonal amniotic fluid-derived stem cells express characteristics of both mesenchymal and neural stem cells. Biol Reprod 2006;74:545-51.
[32] Wang CH, Ciliberti N, Li SH, Szmitko PE, Weisel RD, Fedak PW, et al. Rosiglitazone facilitates angiogenic progenitor cell differentiation toward endothelial lineage: A new paradigm in glitazone pleiotropy. Circulation 2004;109:1392-400.
[33] Oswald J, Boxberger S, Jorgensen B, Feldmann S, Ehninger G, Bornhauser M, et al. Mesenchymal stem cells can be differentiated into endothelial cells in vitro. Stem Cells 2004;22:377-84.
[34] Hida N, Nishiyama N, Miyoshi S, Kira S, Segawa K, Uyama T, et al. Novel cardiac precursor-like cells from human menstrual blood-derived mesenchymal cells. Stem Cells 2008;26;1695-704.
[35] Heng BC, Haider HKh, Sim EK, Cao T, Ng SC. Strategies for directing the differentiation of stem cells into the cardiomyogenic lineage in vitro. Cardiovasc Res 2004;62;34-42.
[36] Uemura R, Xu M, Ahmad N and Ashraf M. Bone marrow stem cells prevent left ventricular remodeling of ischemic heart through paracrine signaling. Circ Res 2006;98:1414-21.
[37] Welikson RE, Kaestner S, Reinecke H and Hauschka SD. Human umbilical vein endothelial cells fuse with cardiomyocytes but do not activate cardiac gene expression. J Mol Cell Cardiol 2006;40:520-8.
[38] Matsubayashi K, Fedak PW, Mickle DA, Weisel RD, Ozawa T, and Li RK. Improved left ventricular aneurysm repair with bioengineered vascular smooth muscle grafts. Circulation 2003;108 Supp 1:II219-25.
[39] Caspi O, Huber I, Kehat I, Habib M, Arbel G, Gepstein A, et al. Transplantation of human embryonic stem cell-derived cardiomyocytes improves myocardial performance in infarcted rat hearts. J Am Coll Cardiol 2007;50:1884-93.
[40] Gandia C, Arminan A, Garcia-Verdugo JM, Lledo E, Ruiz A, Minana MD, et al. Human dental pulp stem cells improve left ventricular function, induce angiogenesis, and reduce infarct size in rats with acute myocardial infarction. Stem Cells 2008;26:638-45.
[41] Toma C, Pittenger MF, Cahill KS, Byrne BJ, and Kessler PD. Human mesenchymal stem cells differentiate to a cardiomyocyte phenotype in the adult murine heart. Circulation 2005;105:93-8.
[42] Davani S, Marandin A, Mersin N, Royer B, Kantelip B, Herve P, et al. Mesenchymal progenitor cells differentiate into an endothelial phenotype, enhance vascular density, and improve heart function in a rat cellular cardiomyoplasty model. Circulation 2003;108 Supp 1:II253-8.
[43] du Toit D, Muller C, Page B, and Louw J. Foetal rat pancreatic transplantation: Posttransplantation development of foetal pancreatic iso- and allografts and suppression of rejection with mycophenolate mofetil (MMF) and cyclosporine based immunesuppression. Microsc Res Tech 1998;43:347-55.
[44] Laflamme MA, Chen KY, Naumova AV, Muskheli V, Fugate JA, Dupras SK, et al. Cardiomyocytes derived from human embryonic stem cells in pro-survival factors enhance function of infracted rat hearts. Nat Biotechnol 2007;25:1015-24.
[45] Lee EJ, Lee HN, Kang HJ, Kim KH, Hur J, Cho HJ, et al. Novel Embryoid Body–Based Method to Derive Mesenchymal Stem Cells from Human Embryonic Stem Cells. Tissue Eng Part A 2010;16:705-15.
[46] Laflamme MA, Gold J, Xu C, Hassanipour M, Rosler E, Police S, Muskheli V, et al. Formation of human myocardium in the rat heart from human embryonic stem cells. Am J Pathol 2005;167:663-71.
[47] Dai W, Hale SL, Martin BJ, Kuang JQ, Dow JS, Wold LE, et al. Allogeneic mesenchymal stem cell transplantation in postinfarcted rat myocardium: Short- and long-term effects. Circulation 2005;112:214-23.
[48] Salameh A, Frenzel C, Boldt A, Rassler B, Glawe I, Schulte J, et al. Subchronic alpha- and beta-adrenergic regulation of cardiac gap junction protein expression. FASEB J 2006;20:365-7.
[49] Wang CC, Chen CH, Hwang SM, Lin WW, Huang CH, Lee WY, et al. Spherically symmetric mesenchymal stromal cell bodies inherent with endogenous extracellular matrices for cellular cardiomyoplasty. Stem Cells 2009;27:724-32.
[50] Wang CC, Chen CH, Lin WW, Hwang SM, Hsieh PC, Lai PH, et al. Direct intramyocardial injection of mesenchymal stem cell sheet fragments improves cardiac functions after infarction. Cardiovasc Res 2008;77:515-24.
[51] Wei HJ, Chen CH, Lee WY, Chiu I, Hwang SM, Lin WW, et al. Bioengineered cardiac patch constructed from multilayered mesenchymal stem cells for myocardial repair. Biomaterials 2008;29:3547-56.
[52] Chen CH, Wei HJ, Lin WW, Chiu I, Hwang SM, Wang CC, et al. Porous tissue grafts sandwiched with multilayered mesenchymal stromal cell sheets induce tissue regeneration for cardiac repair. Cardiovasc Res 2008;80:88-95.
[53] Chao W, Matsui T, Novikov MS, Tao J, Li L, Liu H, et al. Strategic advantages of insulin-like growth factor-I expression for cardioprotection. J Gene Med 2003;5:277-86.
[54] Gnecchi M, Zhang Z, Ni A and Dzau VJ. Paracrine mechanisms in adult stem cell signaling and therapy. Circ Res 2008;103:1204-19.
[55] Yamamoto M, Sakakibara Y, Nishimura K, Komeda M, and Tabata Y. Improved therapeutic efficacy in cardiomyocyte transplantation for myocardial infarction with release system of basic fibroblast growth factor. Artif Organs 2003;27:181-4.
[56] Orlic D, Kajstura J, Chimenti S, Jakoniuk I, Anderson SM, Li B, et al. Bone marrow cells regenerate infarcted myocardium. Nature 2001;410:701–5.
[57] Reffelmann T, Konemann S, Kloner RA. Promise of blood- and bone marrow-derived stem cell transplantation for functional cardiac repair: Putting it in perspective with existing therapy. J Am Coll Cardiol 2009;53:305–8.
[58] Yeh YC, Wei HJ, Lee WY, Yu CL, Chang Y, Hsu LW, et al. Cellular cardiomyoplasty with human amniotic fluid stem cells: In vitro and in vivo studies. Tissue Eng Part A 2010;16:1925-36.
[59] Chen CH, Tsai CC, Chen W, Mi FL, Liang HF, Chen SC, et al. Novel living cell sheet harvest system composed of thermoreversible methylcellulose hydrogels. Biomacromolecules 2006;7:736–43.
[60] Neff JA, Tresco PA, Caldwell KD. Surface modification for controlled studies of cell-ligand interactions. Biomaterials 1999;20:2377–93.
[61] Okada M, Payne TR, Zheng B, Oshima H, Momoi N, Tobita K, et al. Myogenic endothelial cells purified from human skeletal muscle improve cardiac function after transplantation into infarcted myocardium. J Am Coll Cardiol 2008;52:1869–80.
[62] van der Bogt KE, Sheikh AY, Schrepfer S, Hoyt G, Cao F, Ransohoff KJ, et al. Comparison of different adult stem cell types for treatment of myocardial ischemia. Circulation 2008;118:S121–9.
[63] Tan MY, Zhi W, Wei RQ, Huang YC, Zhou KP, Tan B, te al. Repair of infarcted myocardium using mesenchymal stem cell seeded small intestinal submucosa in rabbits. Biomaterials 2009;30:3234–40.
[64] Suzuki K, Murtuza B, Beauchamp JR, Smolenski RT, Varela-Carver A, Fukushima S, et al. Dynamics and mediators of acute graft attrition after myoblast transplantation to the heart. FASEB J 2004;18:1153–5.
[65] Sottile J, Hocking DC, Swiatek PJ. Fibronectin matrix assembly enhances adhesion-dependent cell growth. J Cell Sci. 1998;111:2933–43.
[66] Roubelakis MG, Pappa KI, Bitsika V, Zagoura D, Vlahou A, Papadaki HA, et al. Molecular and proteomic characterization of human mesenchymal stem cells derived from amniotic fluid: comparison to bone marrow mesenchymal stem cells. Stem Cells and Development 2007;16:931–52.
[67] Haque A, Morris ER. Thermogelatin of methylcellulose. Part I: Molecular-structures and processes. Carbohyd Polym 1993;22:161–73.
[68] Haque A, Richardson RK, Morris ER. Thermogelatin of methylcellulose. Part II: Effect of hydroxypropyl substituents. Carbohyd Polym 1993;22:175–86.
[69] Li L, Shan H, Yue CY, Lam YC, Tam KC, Hu X. Thermally induced association and dissociation of methylcellulose in aqueous solutions. Langmuir 2002;18:7291–8.
[70] Xu Y, Li L, Zheng P, Lam YC, Hu X. Controllable gelation of methylcellulose by a salt mixture. Langmuir 2004;20:6134–8.
[71] Terrovitis JV, Smith RR, Marban E. Assessment and optimization of cell engraftment after transplantation into the heart. Circ Research 2010;106:479–94.
[72] Huang NF, Yu J, Sievers R, Li S, Lee RJ. Injectable biopolymers enhance angiogenesis after myocardial infarction. Tissue Eng 2005;11:1860–6.
[73] Dai W, Wold LE, Dow JS, Kloner RA. Thickening of the infarcted wall by collagen injection improves left ventricular function in rats: A novel approach to preserve cardiac function after myocardial infarction. J Am Coll Cardiol 2005;46:714–9.
[74] Singla DK, Lyons GE, Kamp TJ. Transplanted embryonic stem cells following mouse myocardial infarction inhibit apoptosis and cardiac remodeling. Am J Physiol Heart Circ Physiol 2007;293:H1308–14.
[75] Abdulrazzak H, Moschidou D, Jones G, Guillot PV. Biological characteristics of stem cells from foetal, cord blood and extraembryonic tissues. J R Soc Interface 2010 [Epub ahead of print]
[76] Goldstein AS, Juarez TM, Helmke CD, Gustin MC, Mikos AG. Effect of convection on osteoblastic cell growth and function in biodegradable polymer foam scaffolds. Biomaterials 2001;22:1279-88.
[77] Fuchs JR, Terada S, Hannouche D, Ochoa ER, Vacanti JP, Fauza DO. Engineered fetal cartilage: structural and functional analysis in vitro. J Pediatr Surg 2002;37:1720-5.
[78] Martin I, Wendt D, Heberer M. The role of bioreactors in tissue engineering. Trends Biotechnol 2004;22:80-6.
[79] Gooch KJ, Kwon JH, Blunk T, Langer R, Freed LE, Vunjak-Novakovic G. Effects of mixing intensity on tissue-engineered cartilage. Biotechnol Bioeng 2001;72:402-7.
[80] Croughan MS, Hamel JF, Wang DI. Hydrodynamic effects on animal cells grown in microcarrier cultures. Biotechnol Bioeng 1987;29:130-41.
[81] Kang SW, La WG, Kim BS. Open macroporous poly(lactic-co-glycolic Acid) microspheres as an injectable scaffold for cartilage tissue engineering. J Biomater Sci Polym Ed 2009;20:399-409.
[82] Marangoni AG, Tosh SM. On the nature of the maximum gelation temperature in polymer gels. Biophys Chem 2005;113:265-7.
[83] Krebs MD, Sutter KA, Lin AS, Guldberg RE, Alsberg E. Injectable poly(lactic-co-glycolic) acid scaffolds with in situ pore formation for tissue engineering. Acta Biomater. 2009;5:2847-59.
[84] Oh SH, Kim JH, Song KS, Jeon BH, Yoon JH, Seo TB, et al. Peripheral nerve regeneration within an asymmetrically porous PLGA/Pluronic F127 nerve guide conduit. Biomaterials. 2008;29:1601-9.
[85] Melero-Martin JM, Dowling MA, Smith M, Al-Rubeai M. Expansion of chondroprogenitor cells on macroporous microcarriers as an alternative to conventional monolayer systems. Biomaterials 2006;27:2970-9.
[86] Carmeliet P, Jain RK. Angiogenesis in cancer and other diseases. Nature. 2000;407:249-57.
[87] Lin RZ, Chang HY. Recent advances in three-dimensional multicellular spheroid culture for biomedical research. Biotechnol J. 2008;3:1172-84.
[88] Curcio E, Salerno S, Barbieri G, De Bartolo L, Drioli E, Bader A. Mass transfer and metabolic reactions in hepatocyte spheroids cultured in rotating wall gas-permeable membrane system. Biomaterials 2007;28:5487-97.
[89] Lee WY, Chang YH, Yeh YC, Chen CH, Lin KM, Huang CC, et al. The use of injectable spherically symmetric cell aggregates self-assembled in a thermo-responsive hydrogel for enhanced cell transplantation. Biomaterials 2009;30:5505-13.
[90] Glicklis R, Merchuk JC, Cohen S. Modeling mass transfer in hepatocyte spheroids via cell viability, spheroid size, and hepatocellular functions. Biotechnol Bioeng 2004;86:672-80.
[91] Choi SW, Cheong IW, Kim JH, Xia Y. Preparation of uniform microspheres using a simple fluidic device and their crystallization into close-packed lattices. Small 2009;5:454-9.
[92] Shi X, Sun L, Jiang J, Zhang X, Ding W, Gan Z. Biodegradable polymeric microcarriers with controllable porous structure for tissue engineering. Macromol Biosci 2009;9:1211-8.
(此全文限內部瀏覽)
電子全文
摘要
 
 
 
 
第一頁 上一頁 下一頁 最後一頁 top
* *