博碩士論文 104324049 詳細資訊




以作者查詢圖書館館藏 以作者查詢臺灣博碩士 以作者查詢全國書目 勘誤回報 、線上人數:12 、訪客IP:3.137.189.57
姓名 陳麗華(Li-Hua Chen)  查詢紙本館藏   畢業系所 化學工程與材料工程學系
論文名稱 藉由調整水凝膠之表面電荷及軟硬度並嫁接玻連蛋白用以培養人類多功能幹細胞
(Human Pluripotent Stem Cells Cultured on Recombinant Human Vitronectin-Grafted Hydrogels by Adjusting Surface Charge and Elasticity)
相關論文
★ 於不同彈性係數的生醫材料上體外培植造血幹細胞★ 可見光對羊水間葉幹細胞成骨分化之影響
★ 可見光調控神經細胞之基因表現及突觸生長★ 膜純化法及免疫抗體磁珠法用於分離及體外增殖血液幹細胞之研究
★ 人類表皮成長因子的結構穩定性及生物活性測定★ 微環境對羊水間葉幹細胞多功能性基因表現及分化之影響
★ 奈米片段與細胞外基質之改質膜用於臍帶血中造血幹細胞之純化與培養★ 小鼠脂肪幹細胞之膜純化法及細胞外間質對人類脂肪幹細胞影響之研究
★ 利用具有奈米片段與細胞外間質蛋白質的表面改殖材質進行臍帶血造血幹細胞體外培養★ 在不同培養條件下針對大腸癌細胞及組織中癌細胞進行純化、剔除及鑑定之研究
★ 羊水間葉幹細胞培養於細胞外間質改質表面其分化能力及多能性之研究★ 人類脂肪幹細胞的膜純化法與分化能力研究
★ 具有抗藥性之大腸癌細胞株能提高癌胚抗原的表現,但並非是癌症起始細胞★ 羊水間葉幹細胞培養於接枝細胞外間質寡肽與環狀肽具有最佳表面硬度的生醫材料,其增殖能力及多能性之研究
★ 人類體細胞從組成誘導型多能性幹細胞培養在無飼養層上★ 使用不同孔洞大小之耐倫薄膜從脂肪組織中分離及純化人類脂肪幹細胞之研究
檔案 [Endnote RIS 格式]    [Bibtex 格式]    [相關文章]   [文章引用]   [完整記錄]   [館藏目錄]   至系統瀏覽論文 ( 永不開放)
摘要(中) 對於再生醫療與新藥研發而言,人類多能性幹細胞是具有前瞻性的細胞來源,為了避免於臨床應用造成異種汙染,開發化學定義成分的生醫材料來培養人類多能性幹細胞是重要的課題。有幾種生醫材料被發展來培養人類多能性幹細胞並維持其細胞多能性,例如: 塗佈重組玻連蛋白的培養盤以及嫁接從細胞外基質得來的寡肽培養盤。本研究探討合成材料的軟硬度如何影響細胞的繁殖和多能性。
本研究製作嫁接有重組玻連蛋白的高分子水凝膠(polyvinylalcohol-co-itaconic acid)薄膜來研究軟硬度的物理性質如何影響人類胚胎幹細胞 (WA09)的多能性及生長速率。藉由戊二醛交聯劑,我們可以調整高分子水凝膠的軟硬度至10.3 到 30.4 kPa 儲存模數,接著嫁接高分子賴氨酸在水凝膠上作為主鏈並調整表面電荷,最後嫁接重組玻連蛋白於基材上。
本研究調適出最優化的高分子水凝膠軟硬度並嫁接較低濃度的 (5 g/ml)重組玻連蛋白,相較於其他研究需要極高濃度的 (500 g/ml)寡肽。此材料被用來長期培養繁殖人類多能性幹細胞至少10到20代於無異種環境,並藉由螢光染色、胚體的體外分化和畸形瘤的體內生成來鑑定培養10到20代過後的細胞蛋白質是否具有多能性。
關鍵字: 多能性幹細胞、無異種、表面電荷、水凝膠、玻連蛋白
摘要(英)
Abstract
Human pluripotent stem cells (hPSCs) are promising cell source for regenerative medicine and drug discovery. The development of chemically defined biomaterials is necessary for culturing hPSCs for clinical applications without xenogenic contaminants. It is developed that the biomaterials for hPSCs culture to maintain their pluripotency, such as (a) dishes coated with recombinant vitronectin (b) dishes immobilized with oligopeptides derived from extracellular matrices (ECMs). It is investigated that the effect of the elasticity of the synthetic dishes on the pluripotency fate and proliferation of hPSCs in this study.
I developed polyvinylalcohol-co-itaconic acid (PVA-IA) films grafted with recombinant human vitronectin (rVN) to evaluate the physical effect of elasticity of hydrogels grafted with biologically active nanosegments on the pluripotency and proliferation fates of hPSCs (WA09). The PVA-IA hydrogels were prepared with different elasticities ranging from 10.3 to 30.4 kPa storage moduli by controlling the crosslinking time with glutaraldehyde. Subsequently, rhVN was grafted on PVA-IA hydrogels with or without poly-L-lysine main chains for the adjustment of the surface charge of PVA-IA hydrogels.
This study investigates the optimal elasticity of PVA-IA hydrogels grafted with rhVN that is prepared with much less concentration (5 g/ml) compared to the concentration of oligovitronectin (500 g/ml), which was used in previous study for the expansion of hPSCs for a long period of hPSCs culture (10-20 passages) under xeno-free condition. hPSCs cultured on PVA-IA hydrogels grafted with rhVN were evaluated from pluripotent protein expression by immunostaining, embryoid body (EB) formation, and teratoma formation after 10 and 20 passages.
Keywords: pluripotent stem cell, Xeno-free, surface charge, hydrogel, recombinant vitronectin
關鍵字(中) ★ 多能性幹細胞
★ 無異種
★ 表面電荷
★ 水凝膠
★ 玻連蛋白
★ 軟硬度
關鍵字(英) ★ pluripotent stem cell
★ xeno-free
★ surface charge
★ hydrogel
★ recombinant vitronectin
★ elasticity
論文目次
Index of Content
Abstract I
摘要 III
Index of Content IV
Index of Figure VII
Index of Table XII
Chapter 1 Introduction 1
1-1 Stem Cells 1
1-1-1 Human Embryonic Stem Cells (hESCs) 2
1-1-2 Human Induced Pluripotent Stem Cells (hiPSCs) 4
1-2 Expansion of Pluripotent Stem Cell under Xeno-Free Conditions 5
1-2-1 hPSCs Culture on ECM-Coated Surface (2D) 6
1-2-2 hPSCs Culture on Oligopeptide-Immobolized Surface 8
1-2-3 hPSCs Culture on Synthetic Polymer Surface (2D) 10
1-3 Microenvironment Effect on hPSCs 13
1-3-1 The Effect of Biomaterial Elasticity on hPSCs 14
1-3-2 The Effect of Hydrophilicity of Biomaterials on hPSCs Adhesion and Function 21
1-3-3 The Effect of Biomaterial Surface Charges on hPSCs 21
1-3-4 Cell-Cell Interactions 22
1-4 Characterization of hPSCs 22
1-4-1 Colony Formation 24
1-4-2 Alkali Phosphatase Activity 24
1-4-3 Pluripotent Gene Expression 24
1-4-4 Pluripotent Protein Expression 24
1-4-5 Differentiation Ability 25
1-4-6 Immunofluorescence 27
1-5 Cardiomyocyte Differentiation 28
1-5-1 Effects of Elasticity of Biomaterials on The Differentiation of hPSCs into Cardiomyocytes 29
1-5-2 Effects of Biomaterial Surface on The Cardiomyocytes Adhesion 30
1-5-3 Characterization of Cardiomyocytes 31
Chapter2. Material and Methods 36
2-1 Material 36
2-1-1 Cell Line 36
2-1-2 Commercial Coated Dishes 36
2-1-3 Medium and Others 36
2-1-4 Chemical Materials 37
2-1-5 Immunostaining 41
2-2 Cell Culture 42
2-2-1 Preparation of Cross-Linked PVA-IA Hydrogel Dishes 42
2-2-2 Preparation of PVA-IA Hydrogel Dishes Grafted with PLL and rVN 43
2-2-3 hESCs Culturing and Passaging 44
2-2-4 Cryopreservation of hESCs 45
2-2-5 hESCs Thawing 45
2-2-6 Cardiomyocyte Differentiation under Xeno-Free Condition 46
2-3 Characterization of Dish Surface 47
2-3-1 X-ray Photoelectron Spectra 47
2-3-2 Contact Angle Goniometer 47
2-3-3 Zeta Potential Measurements of Hydrogel Surface 48
2-3-4 Zeta-Potential Measurement of Polymer Solution 48
2-4 Characterization of hESCs 49
2-4-1 Cell Density Measurements 49
2-4-2 Expansion Fold and Doubling Time of hPSCs 50
2-4-3 Differentiation Ratio of hPSCs 50
2-4-4 Immunostaining of Cells 51
2-4-5 Embryoid Body Formation 52
2-4-6 Teratoma Formation 54
2-5 Characterization of hESCs-Derived Cardiomyocytes 54
2-5-1 Sarcomere Immunostaining Analysis 54
2-5-2 Flow Cytometry Analysis 56
Chapter 3. Results and Discussion 57
3-1 PVA-IA Films Grafted with 10μg/ml of rVN with Different Elasticities 57
3-1-1 Physical Characterization of PVA-IA Films 57
3-1-2 Characterization of PVA-IA Films Grafted with 10μg/ml of rVN with Different Elasticities 59
3-1-3 Cultivation of hESCs on PVA-IA Films Grafted with 10μg/ml of rVN with Different Elasticities 63
3-2 PVA-IA-24H Films Grafted with Different Concentration of rVN 69
3-2-1 Characterization of PVA-IA-24H Films Grafted with Different Concentration of rVN 69
3-2-2 Cultivation of hESCs on PVA-IA-24H Films Grafted with Different Concentration of rVN 73
3-2-3 The Effect of Hydrophilicity of Biomaterials on hESCs Adhesion and Function 76
3-2-4 The Effect of Biomaterial Surface Charges on hPSCs Culture 80
3-3 PVA-IA Films Grafted with Poly-L-Lysine and 5μg/ml of rVN 85
3-3-1 Characterization of PVA-IA Films Grafted with Poly-L-Lysine and 5μg/ml of rVN 85
3-3-2 Cultivation of hESCs on PVA-IA Films Grafted with PLL and 5μg/ml of rVN with Different Elasticities 87
3-4 Pluripotency Analysis of hESCs After Cultivation on PVA-IA Films Grafted with PLL and rVN Having Different Elasticities for 10 passages. 91
3-4-1 Immunostaining of Pluripotent proteins 91
3-4-2 Differentiation Ability In Vitro – Embryoid Body Formation 98
3-5 The Effect of Elasticity of Biomaterials on The Differentiation of hESCs into Cardiomyocytes 105
Chapter 4. Conclusions 111
Reference 113
Supplementary Data 121
參考文獻

Reference
1. Mahla, R.S., Stem cells applications in regenerative medicine and disease therapeutics. International Journal of Cell Biology, 2016. 2016.
2. Berdasco, M. and M. Esteller, DNA methylation in stem cell renewal and multipotency. Stem cell research & therapy, 2011. 2(5): p. 42.
3. Thomson, J.A., et al., Embryonic stem cell lines derived from human blastocysts. science, 1998. 282(5391): p. 1145-1147.
4. Wichterle, H., et al., Directed differentiation of embryonic stem cells into motor neurons. Cell, 2002. 110(3): p. 385-397.
5. Lu, S.-J., et al., Directed differentiation of red blood cells from human embryonic stem cells. Cellular Programming and Reprogramming: Methods and Protocols, 2010: p. 105-121.
6. Samadikuchaksaraei, A., et al., Derivation of distal airway epithelium from human embryonic stem cells. Tissue Engineering, 2006. 12(4): p. 867-875.
7. Strulovici, Y., et al., Human Embryonic Stem Cells and Gene Therapy. The American Society of Gene Therapy, 2007.
8. N, S., et al., Morula-derived human embryonic stem cells. Reprod Biomed Online, 2004. 9(6): p. 623-629.
9. X, Z., et al., Derivation of human embryonic stem cells from developing and arrested embryos. Stem Cells, 2006. 24(12): p. 2669-2676.
10. Klimanskaya, et al., Human embryonic stem cell lines derived from single blastomeres. Nature, 2006. 444(7118): p. 481-485.
11. Carr, A.-J.F., et al., Development of human embryonic stem cell therapies for age-related macular degeneration. Trends Neurosci, 2013. 36(7): p. 385-395.
12. J, C., et al., Directed differentiation of human embryonic stem cells into functional hepatic cells. Hepatology, 2007. 45(5): p. 1229-1239.
13. A, N., et al., Cotransplantation of human embryonic stem cell-derived neural progenitors and schwann cells in a rat spinal cord contusion injury model elicits a distinct neurogenesis and functional recovery. Cell Transplant, 2012. 21(5): p. 827-843.
14. Strulovici, Y., et al., Human embryonic stem cells and gene therapy. Molecular Therapy, 2007. 15(5): p. 850-866.
15. Takahashi, K., et al., Induction of pluripotent stem cells from adult human fibroblasts by defined factors. cell, 2007. 131(5): p. 861-872.
16. Lin, S.-L., et al., Mir-302 reprograms human skin cancer cells into a pluripotent ES-cell-like state. Rna, 2008. 14(10): p. 2115-2124.
17. Zhou, H., et al., Generation of induced pluripotent stem cells using recombinant proteins. Cell stem cell, 2009. 4(5): p. 381.
18. Higuchi, A., et al., Biomaterials for the feeder-free culture of human embryonic stem cells and induced pluripotent stem cells. Chemical reviews, 2011. 111(5): p. 3021-3035.
19. Loh, K.M. and B. Lim, Stem cells: Close encounters with full potential. Nature, 2013. 502(7469): p. 41-42.
20. Crook, J.M., et al., The Generation of Six Clinical-Grade Human Embryonic Stem Cell Lines. Cell Stem Cell, 2007. 1(5): p. 490-494.
21. Rajala, K., et al., A Defined and Xeno-Free Culture Method Enabling the Establishment of Clinical-Grade Human Embryonic, Induced Pluripotent and Adipose Stem Cells. PLoS ONE, 2010. 5(4): p. .
22. C, X., et al., Basic fibroblast growth factor supports undifferentiated human embryonic stem cell growth without conditioned medium. Stem Cells, 2005. 23(3): p. 315-323.
23. Higuchi, A., et al., Design of polymeric materials for culturing human pluripotent stem cells: Progress toward feeder-free and xeno-free culturing. Progress in Polymer Science, 2014. 39(7): p. 1348-1374.
24. CS, H., P. LM, and L. GA, Matrigel: a complex protein mixture required for optimal growth of cell culture. Proteomics, 2010. 10(9): p. 1886-1890.
25. Abedin, M. and N. King, Diverse evolutionary paths to cell adhesion. Trends Cell Biol, 2010. 20(12): p. 734-742.
26. Fu, X., et al., Establishment of clinically compliant human embryonic stem cells in an autologous feeder-free system. Tissue Engineering Part C, 2011. 17(9): p. 927-937.
27. Chen, G., et al., Chemically defined conditions for human iPSC derivation and culture. nature methods, 2011. 8(8): p. 424-429.
28. AK, K., K. CB, and P. AJ, Extracellular matrix remodeling, integrin expression, and downstream signaling pathways influence the osteogenic differentiation of mesenchymal stem cells on poly(lactide-co-glycolide) substrates. Tissue Eng Part A, 2009. 15(2): p. 273-283.
29. Meng, G., S. Liu, and D.E. Rancourt, Synergistic effect of medium, matrix, and exogenous factors on the adhesion and growth of human pluripotent stem cells under defined, xeno-free conditions. Stem cells and development, 2011. 21(11): p. 2036-2048.
30. Li, J., et al., Impact of vitronectin concentration and surface properties on the stable propagation of human embryonic stem cells. Biointerphases, 2010. 5(3): p. FA132-FA142.
31. Braam, S.R., et al., Recombinant vitronectin is a functionally defined substrate that supports human embryonic stem cell self-renewal via alpha V beta 5 integrin. Stem Cells, 2008. 26(9): p. 2257-2265.
32. Brafman, D.A., et al., Long-term human pluripotent stem cell self-renewal on synthetic polymer surfaces. Biomaterials, 2010. 31(34): p. 9135-9144.
33. Melkoumian, Z., et al., Synthetic peptide-acrylate surfaces for long-term self-renewal and cardiomyocyte differentiation of human embryonic stem cells. Nature biotechnology, 2010. 28(6): p. 606-610.
34. Kolhar, P., et al., Synthetic surfaces for human embryonic stem cell culture. Journal of biotechnology, 2010. 146(3): p. 143-146.
35. Klim, J.R., et al., A defined glycosaminoglycan-binding substratum for human pluripotent stem cells. Nature methods, 2010. 7(12): p. 989-994.
36. Nishishita, N., et al., Generation of virus-free induced pluripotent stem cell clones on a synthetic matrix via a single cell subcloning in the naive state. PLoS One, 2012. 7(6): p. e38389.
37. Chen, Y.-M., et al., Xeno-free culture of human pluripotent stem cells on oligopeptide-grafted hydrogels with various molecular designs. Scientific Reports, 2017. 7: p. 45146.
38. Bagley, J., et al., Extended culture of multipotent hematopoietic progenitors without cytokine augmentation in a novel three-dimensional device. Experimental hematology, 1999. 27(3): p. 496-504.
39. Curran, J.M., R. Chen, and J.A. Hunt, The guidance of human mesenchymal stem cell differentiation in vitro by controlled modifications to the cell substrate. Biomaterials, 2006. 27(27): p. 4783-4793.
40. Hayman, M., et al., Growth of human stem cell-derived neurons on solid three-dimensional polymers. Journal of biochemical and biophysical methods, 2005. 62(3): p. 231-240.
41. Lambshead, J.W., et al., Defining synthetic surfaces for human pluripotent stem cell culture. Cell Regeneration, 2013. 2(1): p. 7.
42. Villa-Diaz, L.G., et al., Synthetic polymer coatings for long-term growth of human embryonic stem cells. Nature biotechnology, 2010. 28(6): p. 581-583.
43. Miao, H.-Q., et al., Modulation of fibroblast growth factor-2 receptor binding, dimerization, signaling, and angiogenic activity by a synthetic heparin-mimicking polyanionic compound. Journal of Clinical Investigation, 1997. 99(7): p. 1565.
44. Chang, C.-W., et al., Engineering cell–material interfaces for long-term expansion of human pluripotent stem cells. Biomaterials, 2013. 34(4): p. 912-921.
45. Yan, Y., et al., Pluripotent stem cell expansion and neural differentiation in 3-D scaffolds of tunable Poisson’s ratio. Acta Biomaterialia, 2017. 49: p. 192-203.
46. Engler, A.J., et al., Matrix elasticity directs stem cell lineage specification. Cell, 2006. 126(4): p. 677-689.
47. Lam, A.T.-L., et al., Cationic surface charge combined with either vitronectin or laminin dictates the evolution of human embryonic stem cells/microcarrier aggregates and cell growth in agitated cultures. Stem cells and development, 2014. 23(14): p. 1688-1703.
48. Benoit, D.S., et al., Small functional groups for controlled differentiation of hydrogel-encapsulated human mesenchymal stem cells. Nature materials, 2008. 7(10): p. 816-823.
49. Higuchi, A., et al., Physical cues of cell culture materials lead the direction of differentiation lineages of pluripotent stem cells. Journal of Materials Chemistry B, 2015. 3(41): p. 8032-8058.
50. Huebsch, N., et al., Harnessing traction-mediated manipulation of the cell/matrix interface to control stem-cell fate. Nature materials, 2010. 9(6): p. 518-526.
51. Leipzig, N.D. and M.S. Shoichet, The effect of substrate stiffness on adult neural stem cell behavior. Biomaterials, 2009. 30(36): p. 6867-6878.
52. Wang, L. and J.P. Stegemann, Thermogelling chitosan and collagen composite hydrogels initiated with β-glycerophosphate for bone tissue engineering. Biomaterials, 2010. 31(14): p. 3976-3985.
53. Guvendiren, M. and J.A. Burdick, Stiffening hydrogels to probe short-and long-term cellular responses to dynamic mechanics. Nature communications, 2012. 3: p. 792.
54. Ghajar, C.M., et al., Mesenchymal stem cells enhance angiogenesis in mechanically viable prevascularized tissues via early matrix metalloproteinase upregulation. Tissue engineering, 2006. 12(10): p. 2875-2888.
55. Wang, L.-S., et al., Injectable biodegradable hydrogels with tunable mechanical properties for the stimulation of neurogenesic differentiation of human mesenchymal stem cells in 3D culture. Biomaterials, 2010. 31(6): p. 1148-1157.
56. Zoldan, J., et al., The influence of scaffold elasticity on germ layer specification of human embryonic stem cells. Biomaterials, 2011. 32(36): p. 9612-9621.
57. Bai, S., et al., Silk scaffolds with tunable mechanical capability for cell differentiation. Acta biomaterialia, 2015. 20: p. 22-31.
58. Murphy, C.M., et al., Mesenchymal stem cell fate is regulated by the composition and mechanical properties of collagen–glycosaminoglycan scaffolds. Journal of the mechanical behavior of biomedical materials, 2012. 11: p. 53-62.
59. Trappmann, B., et al., Extracellular-matrix tethering regulates stem-cell fate. Nature materials, 2012. 11(7): p. 642-649.
60. Muduli, S., et al., Stem cell culture on polyvinyl alcohol hydrogels having different elasticity and immobilized with ECM-derived oligopeptides. Journal of Polymer Engineering.
61. Li, W.-J., et al., Fabrication and characterization of six electrospun poly (α-hydroxy ester)-based fibrous scaffolds for tissue engineering applications. Acta biomaterialia, 2006. 2(4): p. 377-385.
62. Gobaa, S., et al., Artificial niche microarrays for probing single stem cell fate in high throughput. Nature methods, 2011. 8(11): p. 949-955.
63. Liu, S.Q., et al., Biomimetic hydrogels for chondrogenic differentiation of human mesenchymal stem cells to neocartilage. Biomaterials, 2010. 31(28): p. 7298-7307.
64. Xu, Y., et al., Regulating myogenic differentiation of mesenchymal stem cells using thermosensitive hydrogels. Acta biomaterialia, 2015. 26: p. 23-33.
65. Takahashi, Y., M. Yamamoto, and Y. Tabata, Osteogenic differentiation of mesenchymal stem cells in biodegradable sponges composed of gelatin and β-tricalcium phosphate. Biomaterials, 2005. 26(17): p. 3587-3596.
66. Roohani-Esfahani, S.-I., et al., The influence hydroxyapatite nanoparticle shape and size on the properties of biphasic calcium phosphate scaffolds coated with hydroxyapatite–PCL composites. Biomaterials, 2010. 31(21): p. 5498-5509.
67. Huang, X., et al., Osteoinductive‐nanoscaled silk/HA composite scaffolds for bone tissue engineering application. Journal of Biomedical Materials Research Part B: Applied Biomaterials, 2015. 103(7): p. 1402-1414.
68. Mattei, G., et al., Decoupling the role of stiffness from other hydroxyapatite signalling cues in periosteal derived stem cell differentiation. Scientific reports, 2015. 5: p. 10778.
69. Kim, T., et al., Effect of CNT on collagen fiber structure, stiffness assembly kinetics and stem cell differentiation. Materials Science and Engineering: C, 2015. 49: p. 281-289.
70. Discher, D.E., P. Janmey, and Y.-l. Wang, Tissue cells feel and respond to the stiffness of their substrate. Science, 2005. 310(5751): p. 1139-1143.
71. Cigognini, D., et al., Engineering in vitro microenvironments for cell based therapies and drug discovery. Drug discovery today, 2013. 18(21): p. 1099-1108.
72. Buxboim, A., I.L. Ivanovska, and D.E. Discher, Matrix elasticity, cytoskeletal forces and physics of the nucleus: how deeply do cells ‘feel’outside and in? J Cell Sci, 2010. 123(3): p. 297-308.
73. Kobayashi, T. and M. Sokabe, Sensing substrate rigidity by mechanosensitive ion channels with stress fibers and focal adhesions. Current opinion in cell biology, 2010. 22(5): p. 669-676.
74. Reilly, G.C. and A.J. Engler, Intrinsic extracellular matrix properties regulate stem cell differentiation. Journal of biomechanics, 2010. 43(1): p. 55-62.
75. McBeath, R., et al., Cell shape, cytoskeletal tension, and RhoA regulate stem cell lineage commitment. Developmental cell, 2004. 6(4): p. 483-495.
76. Mei, Y., et al., Combinatorial development of biomaterials for clonal growth of human pluripotent stem cells. Nature materials, 2010. 9(9): p. 768-778.
77. Sitterley, G., et al., Attachment and matrix factors. Biofiles (Sigma-Aldrich), 2008. 3: p. 1-28.
78. Wiertz, R., E. Marani, and W. Rutten, Neural cell–cell and cell–substrate adhesion through N-cadherin, N-CAM and L1. Journal of neural engineering, 2011. 8(4): p. 046004.
79. Lakard, S., et al., Adhesion and proliferation of cells on new polymers modified biomaterials. Bioelectrochemistry, 2004. 62(1): p. 19-27.
80. Blau, A., Cell adhesion promotion strategies for signal transduction enhancement in microelectrode array in vitro electrophysiology: an introductory overview and critical discussion. Current opinion in colloid & interface science, 2013. 18(5): p. 481-492.
81. Fan, Y., et al., Facile engineering of xeno-free microcarriers for the scalable cultivation of human pluripotent stem cells in stirred suspension. Tissue Engineering Part A, 2013. 20(3-4): p. 588-599.
82. Bauwens, C.L., et al., Control of human embryonic stem cell colony and aggregate size heterogeneity influences differentiation trajectories. Stem cells, 2008. 26(9): p. 2300-2310.
83. O′Connor, M.D., et al., Alkaline Phosphatase‐Positive Colony Formation Is a Sensitive, Specific, and Quantitative Indicator of Undifferentiated Human Embryonic Stem Cells. Stem cells, 2008. 26(5): p. 1109-1116.
84. Brimble, S.N., et al., Karyotypic stability, genotyping, differentiation, feeder-free maintenance, and gene expression sampling in three human embryonic stem cell lines derived prior to August 9, 2001. Stem cells and development, 2004. 13(6): p. 585-597.
85. Phillips, B.W., et al., Attachment and growth of human embryonic stem cells on microcarriers. Journal of Biotechnology, 2008. 138(1): p. 24-32.
86. Bigdeli, N., et al., Adaptation of human embryonic stem cells to feeder-free and matrix-free culture conditions directly on plastic surfaces. Journal of biotechnology, 2008. 133(1): p. 146-153.
87. Baxter, M.A., et al., Analysis of the distinct functions of growth factors and tissue culture substrates necessary for the long-term self-renewal of human embryonic stem cell lines. Stem Cell Research, 2009. 3(1): p. 28-38.
88. Amit, M., et al., Feeder layer-and serum-free culture of human embryonic stem cells. Biology of reproduction, 2004. 70(3): p. 837-845.
89. Xu, C., et al., Feeder-free growth of undifferentiated human embryonic stem cells. Nature biotechnology, 2001. 19(10): p. 971-974.
90. Sjögren‐Jansson, E., et al., Large‐scale propagation of four undifferentiated human embryonic stem cell lines in a feeder‐free culture system. Developmental Dynamics, 2005. 233(4): p. 1304-1314.
91. Rubart, M. and L.J. Field, Cardiac regeneration: repopulating the heart. Annu. Rev. Physiol., 2006. 68: p. 29-49.
92. Xu, C., et al., Characterization and enrichment of cardiomyocytes derived from human embryonic stem cells. Circulation research, 2002. 91(6): p. 501-508.
93. Mummery, C., et al., Differentiation of human embryonic stem cells to cardiomyocytes. Circulation, 2003. 107(21): p. 2733-2740.
94. Hazeltine, L.B., et al., Temporal impact of substrate mechanics on differentiation of human embryonic stem cells to cardiomyocytes. Acta biomaterialia, 2014. 10(2): p. 604-612.
95. Ribeiro, A.J., et al., Contractility of single cardiomyocytes differentiated from pluripotent stem cells depends on physiological shape and substrate stiffness. Proceedings of the National Academy of Sciences, 2015. 112(41): p. 12705-12710.
96. Patel, A.K., et al., A defined synthetic substrate for serum-free culture of human stem cell derived cardiomyocytes with improved functional maturity identified using combinatorial materials microarrays. Biomaterials, 2015. 61: p. 257-265.
97. Hutcheon, G., et al., Water absorption and surface properties of novel poly (ethylmethacrylate) polymer systems for use in bone and cartilage repair. Biomaterials, 2001. 22(7): p. 667-676.
98. Chen, J., et al., Antibacterial polymeric nanostructures for biomedical applications. Chemical Communications, 2014. 50(93): p. 14482-14493.
99. Stegemann, M., et al., The cell surface of isolated cardiac myocytes—a light microscope study with use of fluorochrome-coupled lectins. Journal of molecular and cellular cardiology, 1990. 22(7): p. 787-803.
100. Kuzmenkin, A., et al., Functional characterization of cardiomyocytes derived from murine induced pluripotent stem cells in vitro. The FASEB Journal, 2009. 23(12): p. 4168-4180.
101. Bhattacharya, S., et al., High efficiency differentiation of human pluripotent stem cells to cardiomyocytes and characterization by flow cytometry. JoVE (Journal of Visualized Experiments), 2014(91): p. e52010-e52010.
102. Sander, V., et al., Isolation and in vitro culture of primary cardiomyocytes from adult zebrafish hearts. Nature protocols, 2013. 8(4): p. 800-809.
103. Carter, N. and M.G. Ormerod, Introduction to the principles of flow cytometry. Flow cytometry: a practical approach, 2000(229): p. 1.
104. Watson, J.V., Introduction to flow cytometry. 2004: Cambridge University Press.
105. Higuchi, A. and T. Iijima, DSC investigation of the states of water in poly (vinyl alcohol) membranes. Polymer, 1985. 26(8): p. 1207-1211.
106. Kumar, S.S., et al., The combined influence of substrate elasticity and surface-grafted molecules on the ex vivo expansion of hematopoietic stem and progenitor cells. Biomaterials, 2013. 34(31): p. 7632-7644.
107. Higuchi, A. and T. Iijima, DSC investigation of the states of water in poly (vinyl alcohol-co-itaconic acid) membranes. Polymer, 1985. 26(12): p. 1833-1837.
108. Lian, X., et al., Directed cardiomyocyte differentiation from human pluripotent stem cells by modulating Wnt/β-catenin signaling under fully defined conditions. Nature protocols, 2013. 8(1): p. 162-175.
109. Placet, V., Characterization of the thermo-mechanical behaviour of Hemp fibres intended for the manufacturing of high performance composites. Composites Part A: Applied Science and Manufacturing, 2009. 40(8): p. 1111-1118.
110. Mano, J., et al., Dynamic mechanical properties of hydroxyapatite-reinforced and porous starch-based degradable biomaterials. Journal of Materials Science: Materials in Medicine, 1999. 10(12): p. 857-862.
111. Higuchi, A., et al., Long-term xeno-free culture of human pluripotent stem cells on hydrogels with optimal elasticity. Scientific reports, 2015. 5: p. 18136.
112. Park, H.-J., et al., Bio-inspired oligovitronectin-grafted surface for enhanced self-renewal and long-term maintenance of human pluripotent stem cells under feeder-free conditions. Biomaterials, 2015. 50: p. 127-139.
指導教授 樋口亞紺(Akon Higuchi) 審核日期 2017-8-22
推文 facebook   plurk   twitter   funp   google   live   udn   HD   myshare   reddit   netvibes   friend   youpush   delicious   baidu   
網路書籤 Google bookmarks   del.icio.us   hemidemi   myshare   

若有論文相關問題,請聯絡國立中央大學圖書館推廣服務組 TEL:(03)422-7151轉57407,或E-mail聯絡  - 隱私權政策聲明