博碩士論文 101324071 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:39 、訪客IP:18.221.151.175
姓名 高士軒(Shih-hsuan Kao)  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 無滋養層培養人類胚胎幹細胞及誘導型多能性幹細胞於 不同軟硬度之奈米片段接枝表面
(Feeder-free Culture of Human ESCs & iPSCs on Dishes Grafted with Cell Adhesion Peptides and Having Different 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★ 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
★ 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
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]   [檢視]  [下載]
  1. 本電子論文使用權限為同意立即開放。
  2. 已達開放權限電子全文僅授權使用者為學術研究之目的,進行個人非營利性質之檢索、閱讀、列印。
  3. 請遵守中華民國著作權法之相關規定,切勿任意重製、散佈、改作、轉貼、播送,以免觸法。

摘要(中) 人類多能性幹細胞擁有特殊的潛在治療應用,可以助於損壞的器官修復及疾病的治療。然而,臨床實驗的測試受限於細胞培養於老鼠纖維母細胞培養層。在未來,必須開發一個能培養人類多能性幹細胞之無外源汙染的材料。無滋養層培養是利用合成生醫材料接枝奈米片段作為幹細胞的培養盤,他可以幫助人類多能性幹細胞 (人類胚胎幹細胞及人類誘導型多能性幹細胞) 繁殖且同時在無滋養層及外源汙染的培養下維持其多能性。
本人研究在無老鼠纖維母細胞之滋養層下培養人類多能性幹細胞在聚乙烯醇-共-衣康酸 (PVA-IA) 表面接枝奈米片段 (KGGPQVTRGDVFTMP [cell-binding domain derived from vitronectin, oligoVN]) 能夠生長且維持其多能性。利用PVA-IA薄膜表面接枝高濃度之寡肽纖粘蛋白且具有最適合軟硬度的培養盤培養人類多能性幹細胞,比較於市面世上販售合成生醫材料 (Synthemax II),細胞表現較高的貼覆率及成長倍率且較低的分化比率。
更者,經10代培養人類胚胎幹細胞 (WA09) 及人類誘導型多能性幹細胞 (HPS0077) 於PVA-IA塗層接枝1000µg/mL的寡肽纖粘蛋白且具有理想軟硬度25.3kPa的表面,其擁有極好的多能性蛋白及分化蛋白表現。此結果指出人類多能性幹細胞可以培養於PVA-IA培養系統且維持多能性。在未來,PVA-IA培養系統可以被用於在無外源汙染及無滋養層的環境下利用人類原始的組織來製造人類誘導型多能性幹細胞。
摘要(英) Human pluripotent stem cells (hPSCs) have significant potential in therapeutic applications for damaged organs or diseases. However, the tentative clinical potential of hPSCs is restricted by the use of mouse embryonic fibroblasts (MEFs) as a feeder layer. It is necessary to develop a suitable culture system without using xeno-contaminated materials for hPSCs culture in future. The feeder-free cultures using synthetic biomaterials having nanosegments as stem cell culture materials can support the propagation of human pluripotent stem cells (hPSCs), human embryonic stem cells (hESCs), and induced pluripotent stem cells (hiPSCs) while maintaining pluripotency in feeder-free and xeno-free cultures.
I investigated that hPSCs could proliferate and keep pluripotenty without usage of a feeder layer of MEFs where hPSCs were cultured on polyvinylalcohol-co-itaconic acid (PVA-IA) grafted with nanosegment (KGGPQVTRGDVFTMP [cell-binding domain derived from vitronectin, oligoVN]). The hPSCs on PVA-IA film grafted with high concentration of oligoVN having optimal elasticity showed higher colony attachment ratio, higher colony expansion fold, and lower differentiation ratio compared to those on commercially availiable synthetic biomaterial (Synethemax II).
Moreover, hESCs (WA09) and hiPSCs (HPS0077) cultured on PVA-IA films grafted with 1000µg/ml of oligo-VN with 25.3kPa elasticity after 10 passage showed excellent pluripotent protein and differentiation protein expression in embryoid boid. This result indicates that hPSCs can be cultured and maintain their pluripotency on PVA-IA culture system. In future, the PVA-IA culture system could be used to generate hiPSCs from primary human tissue cells on xeno-free and feeder-free conditions.
關鍵字(中) ★ 多能性幹細胞 關鍵字(英)
論文目次 Chapter 1: Introduction XIII
1-1 Stem Cells 1
1-1-1 Stem Cell Therapy and Lineage Plasticity 2
1-1-2 Totipotency and Nuclear Transfer 4
1-1-3 Pluripotent Stem Cells 6
1-1-4 Adult Stem Cells 10
1-2 Microenvironment Effect on Human Pluripotent Stem Cell 11
1-2-1 Physical Cues 11
1-2-2 Chemical Effect 13
1-3 Characterization of Pluripotent Stem Cells 44
1-3-1 Colony Formation 46
1-3-2 Alkali Phosphatase Activity 46
1-3-3 Pluripotent Gene Expression 46
1-3-4 Pluripotent Protein Expression 46
1-3-5 Differentiation Ability 47
1-4 Immunofluorescence 49
Chapter 2: Materials and Methods 51
2-1 Material 51
2-1-1 Cell line 51
2-1-2 Chemical 51
2-2 Cell Culture 53
2-2-1 Culture and passage of human pluripotent stem cells 53
2-2-2 Storage of human pluripotent stem cell 54
2-2-3 Thawing frozen stock of human pluripotent stem cell 55
2-3 Immunofluorescence 55
2-4 Colony attachment ratio 57
2-5 Colony differentiation ratio 57
2-6 Alkaline phosphatase live staining assay 58
2-7 Embryoid body formation 59
2-8 Teratoma formation 60
2-9 Preparation of PVA-IA film 61
2-10 Preparation of PVA-IA coating dish grafted with oligopeptide 62
2-11 XPS analysis of dish surface 63
Chapter 3: Result and Discussion 64
3-1 Characterization of PVA-IA film grafted with oligovitronectin by X-ray photoelectron spectroscopy 64
3-2 Cultivation of hPSCs on PVA-IA films grafted with oligo-VN 68
3-2-1 Transfer of hESCs from feeder-layer culture system to feeder-layer free culture system using Matrigel coated dish 68
3-2-2 Characterization of hESCs before cultivation on PVA-IA culture system 70
3-2-3 Cultivation of hESCs on PVA-IA films having different stiffness grafted with oligo-VN with different concentration 72
3-2-4 Characterization of hESCs after cultivation on PVA-IA culture system 84
3-2-5 Cultivation of hiPSCs on PVA-IA films having different stiffness grafted with different concentration of oligo-VN 88
3-2-6 Characterization of hiPSCs after cultivation on PVA-IA culture system 100
3-3 Cultivation of hPSCs on PVA-IA films grafted with different concentration of Oligo-VN having optimal elasticity (25.3kPa) for 20 passages 104
Chapter 4: Conclusion 108
Supplement Data 110
Supplement tables 110
Reference 121
參考文獻 1. Andrews, P.W., et al., Embryonic stem (ES) cells and embryonal carcinoma (EC) cells: opposite sides of the same coin. Biochem Soc Trans, 2005. 33(Pt 6): p. 1526-30.
2. Evans, M.J. and M.H. Kaufman, Establishment in culture of pluripotential cells from mouse embryos. Nature, 1981. 292(5819): p. 154-6.
3. Embryonic and Induced Pluripotent Stem Cells, Sigma-Aldrich, http://www.sigmaaldrich.com/life-science/stem-cell-biology/ipsc.html,6/6/2011 15:05.
4. Thomson, J.A., et al., Embryonic stem cell lines derived from human blastocysts. Science, 1998. 282(5391): p. 1145-7.
5. Mitalipov, S. and D. Wolf, Totipotency, pluripotency and nuclear reprogramming. Adv Biochem Eng Biotechnol, 2009. 114: p. 185-99.
6. Campbell, K.H., et al., Sheep cloned by nuclear transfer from a cultured cell line. Nature, 1996. 380(6569): p. 64-6.
7. Palmarini, M., A veterinary twist on pathogen biology. PLoS Pathog, 2007. 3(2): p. e12.
8. Shiels, P.G., et al., Analysis of telomere lengths in cloned sheep. Nature, 1999. 399(6734): p. 316-7
9. Tachibana, M., et al., Human embryonic stem cells derived by somatic cell nuclear transfer. Cell, 2013. 153(6): p. 1228-38.
10. Chung, Y.G., et al., Human Somatic Cell Nuclear Transfer Using Adult Cells. Cell Stem Cell, 2014.7. Mitalipov, S. and D. Wolf, Totipotency, pluripotency and nuclear reprogramming. Adv Biochem Eng Biotechnol, 2009. 114: p. 185-99.
11. Engler, A.J., et al., Matrix elasticity directs stem cell lineage specification. Cell, 2006. 126(4): p. 677-89.
12. Shih, Y. R. V.; Tseng, K. F.; Lai, H. Y.; Lin, C. H.; Lee, O. K. J.Bone Miner. Res. 2011, 26, 730.
13. Engler, A.J., et al., Matrix elasticity directs stem cell lineage specification. Cell, 2006. 126(4): p. 677-89.
14. Aguilar, H.N. and B.F. Mitchell, Physiological pathways and molecular mechanisms regulating uterine contractility. Hum Reprod Update, 2010. 16(6): p. 725-44
15. Kovács, M., et al., Mechanism of blebbistatin inhibition of myosin II. Journal of Biological Chemistry, 2004. 279(34): p. 35557-35563.
16. Ilic D, Stephenson E, Wood V, Jacquet L, Stevenson D, Petrova A,Kadeva N, Codognotto S, Patel H, Semple M, Cornwell G, OgilvieC, Braude P. Derivation and feeder-free propagation of humanembryonic stem cells under xeno-free conditions. Cytotherapy2012;14:122–8. regulating uterine contractility. Hum Reprod Update, 2010. 16(6): p. 725-44
17. Abraham S, Sheridan SD, Miller B, Rao RR. Stable propagation ofhuman embryonic and induced pluripotent stem cells on decellu-larized human substrates. Biotechnol Prog 2010;26:1126–34.
18. Fu X, Toh WS, Liu H, Lu K, Li M, Cao T. Establishment of clinicallycompliant human embryonic stem cells in an autologous feeder-free system. Tissue Eng C 2011;17:927–37.
19. Meng G, Liu S, Li X, Krawetz R, Rancourt DE. Extracellularmatrix isolated from foreskin fibroblasts supports long-termxeno-free human embryonic stem cell culture. Stem Cells Dev2010;19:547–56.
20. Stelling MP, Lages YM, Tovar AM, Mourao PA, Rehen SK. Matrix-bound heparan sulfate is essential for the growth and pluripotencyof human embryonic stem cells. Glycobiology 2013;23:337–45.
21. Higuchi A, Ling QD, Ko YA, Chang Y, Umezawa A. Biomaterials forthe feeder-free culture of human embryonic stem cells and inducedpluripotent stem cells. Chem Rev 2011;111:3021–35.
22. Rajala K, Hakala H, Panula S, Aivio S, Pihlajamaki H, Suuronen R,Hovatta O, Skottman H. Testing of nine different xeno-free cul-ture media for human embryonic stem cell cultures. Hum Reprod2007;22:1231–8.
23. Manton KJ, Richards S, Van Lonkhuyzen D, Cormack L, Leavesley D,Upton Z. A chimeric vitronectin: IGF-I protein supports feeder-cell-free and serum-free culture of human embryonic stem cells. StemCells Dev 2010;19:1297–305.
24. Zonca Jr MR, Yune PS, Heldt CL, Belfort G, Xie Y. High-throughputscreening of substrate chemistry for embryonic stem cell attach-ment, expansion, and maintaining pluripotency. Macromol Biosci2013;13:177–90.
25. Meng GL, Liu SY, Rancourt DE. Synergistic effect of medium, matrix,and exogenous factors on the adhesion and growth of humanpluripotent stem cells under defined, xeno-free conditions. StemCells Dev 2012;21:2036–48.
26. Higuchi A, Ling QD, Hsu ST, Umezawa A. Biomimetic cell cul-ture proteins as extracellular matrices for stem cell differentiation.Chem Rev 2012;112:4507–40.
27. Gumbiner, B.M., Regulation of cadherin-mediated adhesion in morphogenesis. Nat Rev Mol Cell Biol, 2005. 6(8): p. 622-34.
28. Higuchi, A., et al., Biomaterials for the feeder-free culture of human embryonic stem cells and induced pluripotent stem cells. Chem Rev, 2011. 111(5): p. 3021-35.
29. Ullmann, U., et al., Epithelial-mesenchymal transition process in human embryonic stem cells cultured in feeder-free conditions. Mol Hum Reprod, 2007. 13(1): p. 21-32.
30. Nagaoka, M., et al., Culture of human pluripotent stem cells using completely defined conditions on a recombinant E-cadherin substratum. Bmc Developmental Biology, 2010. 10.
31. Dedhar, S., Cell-substrate interactions and signaling through ILK. Current Opinion in Cell Biology, 2000. 12(2): p. 250-256.
32. Pashuck, E.T. and M.M. Stevens, Designing Regenerative Biomaterial Therapies for the Clinic. Science Translational Medicine, 2012. 4(160).
33. Stephenson, E., et al., Derivation and propagation of human embryonic stem cell lines from frozen embryos in an animal product-free environment. Nature Protocols, 2012. 7(7): p. 1366-1381.
34. Li, Z.S., et al., Feeder-free self-renewal of human embryonic stem cells in 3D porous natural polymer scaffolds. Biomaterials, 2010. 31(3): p. 404-412.
35. Liu, Y.X., et al., Modified Hyaluronan Hydrogels Support the Maintenance of Mouse Embryonic Stem Cells and Human Induced Pluripotent Stem Cells. Macromolecular Bioscience, 2012. 12(8): p. 1034-1042.
36. Li, Z.S., et al., Feeder-free self-renewal of human embryonic stem cells in 3D porous natural polymer scaffolds. Biomaterials, 2010. 31(3): p. 404-412.
37. Siti-Ismail, N., et al., The benefit of human embryonic stem cell encapsulation for prolonged feeder-free maintenance. Biomaterials, 2008. 29(29): p. 3946-3952.
38. Klim, J.R., et al., A defined glycosaminoglycan-binding substratum for human pluripotent stem cells. Nature Methods, 2010. 7(12): p. 989-U72.
39. Gerecht, S., et al., Hyaluronic acid hydrogel for controlled self-renewal and differentiation of human embryonic stem cells. Proc Natl Acad Sci U S A, 2007. 104(27): p. 11298-303.
40. Nandivada, H., et al., Fabrication of synthetic polymer coatings and their use in feeder-free culture of human embryonic stem cells. Nature Protocols, 2011. 6(7): p. 1037-1043.
41. Mei, Y., et al., Combinatorial development of biomaterials for clonal growth of human pluripotent stem cells. Nature Materials, 2010. 9(9): p. 768-778.
42. Mahlstedt, M.M., et al., Maintenance of Pluripotency in Human Embryonic Stem Cells Cultured on a Synthetic Substrate in Conditioned Medium. Biotechnology and Bioengineering, 2010. 105(1): p. 130-140.
43. Nie, Y., et al., Scalable Culture and Cryopreservation of Human Embryonic Stem Cells on Microcarriers. Biotechnology Progress, 2009. 25(1): p. 20-31.
44. Kim, S., et al., A novel culture technique for human embryonic stem cells using porous membranes. Stem Cells, 2007. 25(10): p. 2601-2609.
45. 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.
46. 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.
47. Brafman, D.A., et al., Long-term human pluripotent stem cell self-renewal on synthetic polymer surfaces. Biomaterials, 2010. 31(34): p. 9135-9144.
48. Nandivada, H., et al., Fabrication of synthetic polymer coatings and their use in feeder-free culture of human embryonic stem cells. Nature Protocols, 2011. 6(7): p. 1037-1043.
49. Irwin, E.E., et al., Engineered polymer-media interfaces for the long-term self-renewal of human embryonic stem cells. Biomaterials, 2011. 32(29): p. 6912-6919.
50. Ross, A.M., et al., Synthetic substrates for long-term stem cell culture. Polymer, 2012. 53(13): p. 2533-2539.
51. Zhang, R., et al., A thermoresponsive and chemically defined hydrogel for long-term culture of human embryonic stem cells. Nature Communications, 2013. 4.
52. Higuchi, A., et al., Photon-modulated changes of cell attachments on poly(spiropyran-co-methyl methacrylate) membranes. Biomacromolecules, 2004. 5(5): p. 1770-4.
53. Higuchi, A., et al., Temperature-dependent cell detachment on Pluronic gels. Biomacromolecules, 2005. 6(2): p. 691-6.
54. Tamura, A., et al., Temperature-responsive poly(N-isopropylacrylamide)-grafted microcarriers for large-scale non-invasive harvest of anchorage-dependent cells. Biomaterials, 2012. 33(15): p. 3803-12.
55. Saito, T., et al., Reversal of Diabetes by the Creation of Neo-Islet Tissues Into a Subcutaneous Site Using Islet Cell Sheets. Transplantation, 2011. 92(11): p. 1231-1236.
56. Wei, H., et al., Thermo-sensitive polymeric micelles based on poly(N-isopropylacrylamide) as drug carriers. Progress in Polymer Science, 2009. 34(9): p. 893-910.
57. Carlson, A.L., et al., Microfibrous substrate geometry as a critical trigger for organization, self-renewal, and differentiation of human embryonic stem cells within synthetic 3-dimensional microenvironments. Faseb Journal, 2012. 26(8): p. 3240-3251.
58. Kraehenbuehl, T.P., R. Langer, and L.S. Ferreira, Three-dimensional biomaterials for the study of human pluripotent stem cells. Nat Methods, 2011. 8(9): p. 731-6.
59. 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 Dev, 2012. 21(11): p. 2036-48.
60. 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.
61. Rodin, S., et al., Long-term self-renewal of human pluripotent stem cells on human recombinant laminin-511. Nature Biotechnology, 2010. 28(6): p. 611-U102.
62. Li, J.A., 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.
63. 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).
64. Kim, B.S., et al., Design of artificial extracellular matrices for tissue engineering. Progress in Polymer Science, 2011. 36(2): p. 238-268.
65. 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-U95.
66. Hoffman, L.M. and M.K. Carpenter, Characterization and culture of human embryonic stem cells. Nature Biotechnology, 2005. 23(6): p. 699-708.
67. Kolhar, P., et al., Synthetic surfaces for human embryonic stem cell culture. Journal of Biotechnology, 2010. 146(3): p. 143-146.
68. Klim, J.R., et al., A defined glycosaminoglycan-binding substratum for human pluripotent stem cells. Nature Methods, 2010. 7(12): p. 989-U72.
69. 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 Dev, 2012. 21(11): p. 2036-48.
70. Phillips, B., et al., Attachment and growth of human embryonic stem cells on microcarriers (vol 138, pg 24, 2008). Journal of Biotechnology, 2009. 139(2): p. 194-194.
71. Janssens, S., et al., Autologous bone marrow-derived stem-cell transfer in patients with ST-segment elevation myocardial infarction: double-blind, randomised controlled trial. Lancet, 2006. 367(9505): p. 113-121.
72. Serra, M., et al., Microencapsulation technology: a powerful tool for integrating expansion and cryopreservation of human embryonic stem cells. PLoS One, 2011. 6(8): p. e23212.
73. Steiner, D., et al., Derivation, propagation and controlled differentiation of human embryonic stem cells in suspension. Nat Biotechnol, 2010. 28(4): p. 361-4.
74. Amit, M., et al., Suspension Culture of Undifferentiated Human Embryonic and Induced Pluripotent Stem Cells. Stem Cell Reviews and Reports, 2010. 6(2): p. 248-259.
75. Olmer, R., et al., Long term expansion of undifferentiated human iPS and ES cells in suspension culture using a defined medium. Stem Cell Research, 2010. 5(1): p. 51-64.
76. Zweigerdt, R., et al., Scalable expansion of human pluripotent stem cells in suspension culture. Nature Protocols, 2011. 6(5): p. 689-700.
77. Amit, M., et al., Dynamic suspension culture for scalable expansion of undifferentiated human pluripotent stem cells. Nature Protocols, 2011. 6(5): p. 572-579.
78. Larijani, M.R., et al., Long-Term Maintenance of Undifferentiated Human Embryonic and Induced Pluripotent Stem Cells in Suspension. Stem Cells and Development, 2011. 20(11): p. 1911-1923.
79. Marinho, P.A.N., et al., Xeno-Free Production of Human Embryonic Stem Cells in Stirred Microcarrier Systems Using a Novel Animal/Human-Component-Free Medium. Tissue Engineering Part C-Methods, 2013. 19(2): p. 146-155.
80. Chen, A.K., et al., Critical microcarrier properties affecting the expansion of undifferentiated human embryonic stem cells. Stem Cell Res, 2011. 7(2): p. 97-111.
81. Fernandes, A.M., et al., Successful scale-up of human embryonic stem cell production in a stirred microcarrier culture system. Brazilian Journal of Medical and Biological Research, 2009. 42(6): p. 515-522.
82. Serra, M., et al., Improving expansion of pluripotent human embryonic stem cells in perfused bioreactors through oxygen control. Journal of Biotechnology, 2010. 148(4): p. 208-215.
83. Oh, S.K.W., et al., Long-term microcarrier suspension cultures of human embryonic stem cells. Stem Cell Research, 2009. 2(3): p. 219-230.
84. Bardy, J., et al., Microcarrier Suspension Cultures for High-Density Expansion and Differentiation of Human Pluripotent Stem Cells to Neural Progenitor Cells. Tissue Engineering Part C-Methods, 2013. 19(2): p. 166-180.
85. Storm, M.P., et al., Three-Dimensional Culture Systems for the Expansion of Pluripotent Embryonic Stem Cells. Biotechnology and Bioengineering, 2010. 107(4): p. 683-695.
86. Lock, L.T. and E.S. Tzanakakis, Expansion and Differentiation of Human Embryonic Stem Cells to Endoderm Progeny in a Microcarrier Stirred-Suspension Culture. Tissue Engineering Part A, 2009. 15(8): p. 2051-2063.
87. Heng, B.C., et al., Translating Human Embryonic Stem Cells from 2-Dimensional to 3-Dimensional Cultures in a Defined Medium on Laminin- and Vitronectin-Coated Surfaces. Stem Cells and Development, 2012. 21(10): p. 1701-1715.
88. Nie, Y., et al., Scalable Culture and Cryopreservation of Human Embryonic Stem Cells on Microcarriers. Biotechnology Progress, 2009. 25(1): p. 20-31
89. Tamura, A., et al., Temperature-responsive poly(N-isopropylacrylamide)-grafted microcarriers for large-scale non-invasive harvest of anchorage-dependent cells. Biomaterials, 2012. 33(15): p. 3803-12.
90. Ko, D.Y., et al., Recent progress of in situ formed gels for biomedical applications. Progress in Polymer Science, 2013. 38(3-4): p. 672-701.
91. Lee, K.Y. and D.J. Mooney, Alginate: properties and biomedical applications. Prog Polym Sci, 2012. 37(1): p. 106-126.
92. Huang, X.B., et al., Microenvironment of alginate-based microcapsules for cell culture and tissue engineering. Journal of Bioscience and Bioengineering, 2012. 114(1): p. 1-8.
93. Siti-Ismail, N., et al., The benefit of human embryonic stem cell encapsulation for prolonged feeder-free maintenance. Biomaterials, 2008. 29(29): p. 3946-3952.
94. Wilson, J.L. and T.C. McDevitt, Stem cell microencapsulation for phenotypic control, bioprocessing, and transplantation. Biotechnology and Bioengineering, 2013. 110(3): p. 667-682.
95. Jang, M., et al., A feeder-free, defined three-dimensional polyethylene glycol-based extracellular matrix niche for culture of human embryonic stem cells. Biomaterials, 2013. 34(14): p. 3571-3580.
96. Lutolf, M.R., et al., Repair of bone defects using synthetic mimetics of collagenous extracellular matrices. Nature Biotechnology, 2003. 21(5): p. 513-518.
97. Higuchi, A., et al., Physical cues of biomaterials guide stem cell differentiation fate. Chem Rev, 2013. 113(5): p. 3297-328.
98. Ameen, C., et al., Human embryonic stem cells: current technologies and emerging industrial applications. Crit Rev Oncol Hematol, 2008. 65(1): p. 54-80.
99. 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.
100. Thomson, J.A., et al., Embryonic stem cell lines derived from human blastocysts. Science, 1998. 282(5391): p. 1145-7.
101. Takahashi, K., et al., Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell, 2007. 131(5): p. 861-72.
102. 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 Dev, 2004. 13(6): p. 585-97.
103. Xu, C., et al., Basic fibroblast growth factor supports undifferentiated human embryonic stem cell growth without conditioned medium. Stem Cells, 2005. 23(3): p. 315-23.
104. Phillips, B.W., et al., Attachment and growth of human embryonic stem cells on microcarriers. J Biotechnol, 2008. 138(1-2): p. 24-32.
105. Bigdeli, N., et al., Adaptation of human embryonic stem cells to feeder-free and matrix-free culture conditions directly on plastic surfaces. J Biotechnol, 2008. 133(1): p. 146-53.
106. 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 Res, 2009. 3(1): p. 28-38.
107. Amit, M., et al., Feeder layer- and serum-free culture of human embryonic stem cells. Biol Reprod, 2004. 70(3): p. 837-45.
108. 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. 1109-16.
109. Kokubu, F., et al., Diverse organization of immunoglobulin VH gene loci in a primitive vertebrate. EMBO J, 1988. 7(11): p. 3413-22.
110. Pera, M.F., B. Reubinoff, and A. Trounson, Human embryonic stem cells. J Cell Sci, 2000. 113 ( Pt 1): p. 5-10.
111. Andrews, P.W., et al., Two monoclonal antibodies recognizing determinants on human embryonal carcinoma cells react specifically with the liver isozyme of human alkaline phosphatase. Hybridoma, 1984. 3(1): p. 33-9.
112. Amit, M., et al., Feeder layer- and serum-free culture of human embryonic stem cells. Biol Reprod, 2004. 70(3): p. 837-45.
113. Yamanaka, S., et al., Pluripotency of embryonic stem cells. Cell Tissue Res, 2008. 331(1): p. 5-22.
114. Xu, C., et al., Feeder-free growth of undifferentiated human embryonic stem cells. Nat Biotechnol, 2001. 19(10): p. 971-4.
115. Harkness, L., et al., Isolation and differentiation of chondrocytic cells derived from human embryonic stem cells using dlk1/FA1 as a novel surface marker. Stem Cell Rev, 2009. 5(4): p. 353-68.
116. Sjogren-Jansson, E., et al., Large-scale propagation of four undifferentiated human embryonic stem cell lines in a feeder-free culture system. Dev Dyn, 2005. 233(4): p. 1304-14.
117. Ameen, C., et al., Human embryonic stem cells: current technologies and emerging industrial applications. Crit Rev Oncol Hematol, 2008. 65(1): p. 54-80.
118. Richards, M., et al., Human feeders support prolonged undifferentiated growth of human inner cell masses and embryonic stem cells. Nat Biotechnol, 2002. 20(9): p. 933-6.
119. Peiffer, I., et al., Use of xenofree matrices and molecularly-defined media to control human embryonic stem cell pluripotency: effect of low physiological TGF-beta concentrations. Stem Cells Dev, 2008. 17(3): p. 519-33.
120. Miyazaki, T., et al., Recombinant human laminin isoforms can support the undifferentiated growth of human embryonic stem cells. Biochem Biophys Res Commun, 2008. 375(1): p. 27-32.
121. Zhou, J., et al., mTOR supports long-term self-renewal and suppresses mesoderm and endoderm activities of human embryonic stem cells. Proc Natl Acad Sci U S A, 2009. 106(19): p. 7840-5.
122. Su, Z., et al., Differentiation of human embryonic stem cells into immunostimulatory dendritic cells under feeder-free culture conditions. Clin Cancer Res, 2008. 14(19): p. 6207-17.
123. Li, Z., et al., Feeder-free self-renewal of human embryonic stem cells in 3D porous natural polymer scaffolds. Biomaterials, 2010. 31(3): p. 404-12.
124. Braam, S.R., et al., Recombinant vitronectin is a functionally defined substrate that supports human embryonic stem cell self-renewal via alphavbeta5 integrin. Stem Cells, 2008. 26(9): p. 2257-65.
125. Rosler, E.S., et al., Long-term culture of human embryonic stem cells in feeder-free conditions. Dev Dyn, 2004. 229(2): p. 259-74.
126. Odell, I.D. and D. Cook, Immunofluorescence techniques. J Invest Dermatol, 2013. 133(1): p. e4.
127. A. Higuchi, Q.D. Ling, S. Kumar, M. Munusamy, A.A. Alarfajj, A. Umezawa, et al.
Design of polymeric materials for culturing human pluripotent stem cells: progress toward feeder-free and xeno-free culturing Prog Polym Sci, 39 (7) (2014), pp. 1348–1374
128. Takahashi, K. and S. Yamanaka, Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell, 2006. 126(4): p. 663-76.
指導教授 樋口亞紺(Akon Higuchi) 審核日期 2015-7-30
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