博碩士論文 105324019 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:2 、訪客IP:3.15.190.144
姓名 潘智賢(Chih-Hsien Pan)  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 人類胚胎幹細胞在無滋養層及無異種條件的培養下於帶有生長因子的表面進行培養
(Human embryonic stem cells culture on hydrogels immobilized with human FGF-2)
相關論文
★ 鈣鈦礦膜缺陷控制及製備高效率鈣鈦礦太陽能電池★ 噻吩并[3,4-c]吡咯-4, 6-二酮(TPD)之鈀催化直接碳氫鍵芳香環化反應之研究: 以高步驟經濟效益合成策略製備含TPD之功能性π-共軛小分子
★ 銅觸媒催化之直接碳氫鍵芳香環化反應:以高效率低成本之新合成策略製備含噻吩并[3,4-c]吡咯-4, 6 -二酮(TPD)之功能性有機材料★ 芳香雜環碘化物之水相自身耦合反應:有機光電材料重要前驅物之綠色化學合成法之研究
★ 鈷觸媒催化之芳香雜環溴化物還原性烷基化反應: 以一鍋化反應製備烷基噻吩、呋喃、硒吩與吡咯之有機光電材料重要前驅物★ 利用碳氫鍵芳香環化反應高效率合成小分子有機半導體材料之末端基與其在染料敏化太陽能電池之應用
★ 銅催化之碳氫鍵芳香環化反應於染料敏化太陽能電池之綠色合成與應用★ 利用交叉脫氫耦合反應快速製備各式D-π-A有機染料分子:C-H/C-H合成法研究與其在染料敏化太陽能電池之應用
★ 鈀催化之碳氫鍵活化反應: 發展省步驟且具位向選擇性新合成途徑 快速製備有機光電材料重要之π-A-π結構★ 發展功能性π共軛小分子之綠色合成方法學及其於太陽能電池之應用
★ 利用碳氫鍵芳香環化反應一步合成含EDOT電洞傳輸材料及其於鈣鈦礦太陽能電池之應用★ 碳氫鍵活化反應於鈣鈦礦太陽能電池之應用:高效能電洞傳輸材料合成捷徑之研究
★ 結合C-H/C-Br與C-H/C-H反應快速合成快速合成有機光敏化劑並探討內部拉電子基對於有機光敏化劑並探討內部拉電子基對於 有機光敏化劑並探討內部拉電子基對於 有機光敏化劑並探討內部拉電子基對於 有機光敏化劑並探討內部拉電子基對於 有機光敏化劑並探討內部拉電子基對於染料敏化太陽能電池效率之染料敏化太陽能電池效率之染料敏化太陽能電池效率之染料敏化太陽能電池效率之影響★ 一鍋化連續式碳氫鍵芳香環化反應於染料敏化太陽能電池與鈣鈦礦太陽能電池之應用
★ 利用反應條件最佳化之碳-氫/碳-溴合成策略快速製備以并三?吩為核心結構之電洞傳輸材料★ 含?唑與吩??之電洞傳輸材料於鈣鈦礦太陽能電池之應用:發展碳氫鍵芳香環化之省步驟合成法
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) 人類多功能幹細胞再生醫跟再生醫學領域來說是一種非常有吸引力的的細胞,因為它們具有多能性以至於可以去分化成不同的人體細胞。所以如何去培養人類胚胎幹細胞已經成為一個熱門的主題。通常在多數的細胞研究中需要換培養液,且培養液中的生長因子需要儲存在非常低溫的冰箱下不然會降解跟變形。而降解跟變形的生長因子是造成培養人類多功能幹細胞的培養的成本很高的原因之一。 
在這個研究中把血管生成之纖維母細胞生長因子(FGF-2)嫁接在培養皿上有兩個目的。其一是讓細胞培養中減少培養液中FGF-2的用量;其二是讓生長因子嫁接在表面以至於可以使用比培養液懸浮的生長因子單位密度還要低還可以維持細胞的多能性。那為了確保實驗室成功的我們使用另外一種沒有FGF-2的培養液去培養細胞。目前使用兩種水凝膠來當作基底,嫁接了半胱氨酸與寡?。其中生長因子會與交聯劑鍵結後與半胱氨酸鍵結於水凝膠表面上。在目前的實驗中我們已經可以維持細胞的多能性且讓細胞可以培養超過代。我們堅信我們開發的材料表面應該是可以讓人類多功能幹細胞在培養中保持多能性。
短期目標是能讓人類多功能幹細胞能在生長因子嫁接的表面上維持多能性,最終目標是能夠嫁接不同生長因子促進人類多功能幹細胞分化成特定的細胞。
摘要(英) Human embryonic stem cells (hESCs) are an attractive prospect for regenerative medicine and tissue engineering because of their pluripotency to differentiate into different types of cells. Therefore, how to culture hPSCs has been to consider a very critical issues. Typically, the culture medium of hESCs contains growth factors such as FGF-2 and TGF-β1, which should be stored at -20 ~ -80 degree to prevent denaturation of the growth factors. This process is leading to the high cost of hESC culture medium.
In this experiment, we designed hESC culture dishes immobilized with FGF-2 to reduce the usage of FGF-2 in the culture medium. PVA-IA and CMC hydrogels were grafted with cysteine, which enables to graft the oligopeptide (VN2C) and growth factor (FGF-2) via crosslinker of PEG-SPDP. hESCs (H9) are expected to proliferate and maintain their pluripotency on modified hydrogel surface grafted with FGF-2 in the medium without using FGF-2. To verify this idea, hESCs were cultured on FGF-2 immobilized surface in Essential 6 cell culture medium plus TGF-β1, which corresponds to Essential 8 cell culture medium minus FGF-2. We added Essential 8 medium at first day to stabilize the hESCs to attach on the surface. hESCs culture on CMC-VN2C-FGF surface were found to keep their pluripotency after long term cultivation (>5 passages).
It should be a promising surface, which is immobilized growth factor on cell culture dishes for reducing usage of growth factor such as FGF-2 in the culture medium of hESCs as well as human induced pluripotent stem cells (hiPSCs). The next step is to immobilized the growth factors on the surface to control the hESCs differentiation into specific lineages of the cells.
?
關鍵字(中) ★ 人類胚胎幹細胞
★ 培養
★ 水凝膠
★ 多能性
★ 生長因子
關鍵字(英) ★ human embryonic stem cell
★ culture
★ hydrogel
★ pluripotency
★ growth factor
論文目次 ABSTRACT
摘要 VI
INDEX OF CONTENT VII
INDEX OF FIGURE X
INDEX OF TABLE XV
Chapter 1 Introduction 1
1-1 Regenerative medicine 1
1-1-1 Stem cell therapy 2
1-2 Stem Cells 3
1-2-1 Differentiation potency of stem cell 4
1-3 Human pluripotent stem cells (hPSCs) 6
1-3-1 Human embryonic stem cells (hESCs) 6
1-3-2 Human induced pluripotent stem cells (hiPSCs) 7
1-3-3 Limitation of pluripotent stem cells in clinical trials 7
1-4 Cultivation of human pluripotent stem cells (hPSCs) 9
1-4-1 Extracellular matrix (ECM) coated dishes 10
1-4-2 Oligopeptide-grafted surface 13
1-5 Microenvironment effect for hPSCs culture 15
1-5-1 Elasticity of biomaterials for hPSCs cultivation 16
1-5-2 The effect of hydrophilicity of biomaterials on hPSCs adhesion and function 18
1-5-3 Cell-cell interaction and cell-ECM interaction 19
1-5-4 Soluble factors for hPSCs culture 20
1-6 Characteristic of hPSCs 27
1-6-1 First screening—Colony forming morphology 27
1-6-2 Second screening—Alkaline phosphatase (ALP) activity 28
1-6-3 Second screening—Expression of pluripotent proteins and genes 28
1-6-4 Third screening—Differentiation ability of hPSCs 28
1-7 Goal of this study 29
2-1 Materials 33
2-1-1 Cell line 33
2-1-2 Cell culture media, components, and buffer 33
2-1-3 Surface modification of hydrogels 35
2-1-4 Immunofluorescence staining of hESCs for pluripotency and differentiation assay 37
2-2 Method 39
2-2-1 Preparation of FGF-2 immobilized surface 39
2-2-2 hESCs culture and passage procedure 44
2-2-3 Characterization of FGF-2 immobilized surfaces 46
2-2-4 Characterization of cells growth, pluripotency expression, and differentiation ability in vitro and in vivo 47
Chapter 3 Results and Discussion 53
3-1 Screening of the optimal concentration of cysteine grafted on PVA-IA hydrogels immobilized FGF-2 53
3-1-1 Characterization of PVA-IA hydrogels immobilized FGF-2 with different concentration of cysteine by XPS analysis 54
3-1-2 Characterization of PVA-IA hydrogels immobilized FGF-2 with different concentration of cysteine by ELISA 57
3-1-3 hESCs cultured on PVA-IA hydrogels immobilized FGF-2 with different concentration of cysteine 59
3-2 PVA-IA hydrogels immobilized FGF-2 with increasing the FGF-2 concentration 64
3-2-1 Characterization of PVA-IA hydrogels immobilized FGF-2 with increasing FGF-2 concentration during conjugation reaction by XPS 65
3-2-2 ELISA of PVA-IA hydrogels immobilized FGF-2 with increasing FGF-2 concentration during conjugation reaction 67
3-2-3 hESCs cultivation on PVA-IA hydrogels immobilized FGF-2 with increasing FGF-2 concentration during conjugation reaction 70
3-3 o-Carboxylmethyl chitosan (CMC)-based hydrogels immobilized with FGF-2 75
3-3-1 Water contact angle measurements of CMC-based hydrogels grafted with FGF-2 76
3-3-2 XPS analysis of CMC-based hydrogels immobilized with FGF-2 78
3-3-3 FGF-2 evaluation of CMC-based hydrogels immobilized with FGF-2 by ELISA 84
3-3-4 hESCs cultivation on CMC-based hydrogels immobilized FGF-2 86
3-3-5 Pluripotent analysis of hESCs after long term cultivation on CMC-based hydrogels grafted with FGF-2 98
Chapter 4 Conclusion 106
References 108
參考文獻 1. Lysaght, M.J. and J. Crager, Origins. Tissue Eng Part A, 2009. 15(7): p. 1449-50.
2. Kaiser, L.R., The future of multihospital systems. Top Health Care Financ, 1992. 18(4): p. 32-45.
3. Bajaj, P., et al., 3D biofabrication strategies for tissue engineering and regenerative medicine. Annu Rev Biomed Eng, 2014. 16: p. 247-76.
4. Mao, A.S. and D.J. Mooney, Regenerative medicine: current therapies and future directions. Proceedings of the National Academy of Sciences, 2015. 112(47): p. 14452-14459.
5. Falanga, V. and M. Sabolinski, A bilayered living skin construct (APLIGRAF) accelerates complete closure of hard-to-heal venous ulcers. Wound Repair Regen, 1999. 7(4): p. 201-7.
6. Riazi, A.M., S.Y. Kwon, and W.L. Stanford, Stem cell sources for regenerative medicine, in Stem Cells in Regenerative Medicine. 2009, Springer. p. 55-90.
7. Schwartz, S.D., et al., Embryonic stem cell trials for macular degeneration: a preliminary report. The Lancet, 2012. 379(9817): p. 713-720.
8. Strulovici, Y., et al., Human embryonic stem cells and gene therapy. Molecular Therapy, 2007. 15(5): p. 850-866.
9. Trounson, A. and C. McDonald, Stem cell therapies in clinical trials: progress and challenges. Cell stem cell, 2015. 17(1): p. 11-22.
10. Higuchi, A., et al., Stem Cell Therapies for Reversing Vision Loss. Trends in biotechnology, 2017.
11. Song, W.K., et al., Treatment of macular degeneration using embryonic stem cell-derived retinal pigment epithelium: preliminary results in Asian patients. Stem Cell Reports, 2015. 4(5): p. 860-72.
12. Mandai, M., et al., Autologous induced stem-cell–derived retinal cells for macular degeneration. New England Journal of Medicine, 2017. 376(11): p. 1038-1046.
13. Chen, S.-l., et al., Effect on left ventricular function of intracoronary transplantation of autologous bone marrow mesenchymal stem cell in patients with acute myocardial infarction. American Journal of Cardiology, 2004. 94(1): p. 92-95.
14. Kang, H.-J., et al., Effects of intracoronary infusion of peripheral blood stem-cells mobilised with granulocyte-colony stimulating factor on left ventricular systolic function and restenosis after coronary stenting in myocardial infarction: the MAGIC cell randomised clinical trial. The Lancet, 2004. 363(9411): p. 751-756.
15. Wollert, K.C., et al., Intracoronary autologous bone-marrow cell transfer after myocardial infarction: the BOOST randomised controlled clinical trial. The Lancet, 2004. 364(9429): p. 141-148.
16. Janssens, S., et al., Autologous bone marrow-derived stem-cell transfer in patients with ST-segment elevation myocardial infarction: double-blind, randomised controlled trial. The Lancet, 2006. 367(9505): p. 113-121.
17. Voltarelli, J.C., et al., Autologous nonmyeloablative hematopoietic stem cell transplantation in newly diagnosed type 1 diabetes mellitus. Jama, 2007. 297(14): p. 1568-1576.
18. Yoon, S.H., et al., Complete spinal cord injury treatment using autologous bone marrow cell transplantation and bone marrow stimulation with granulocyte macrophage?colony stimulating factor: phase I/II clinical trial. Stem cells, 2007. 25(8): p. 2066-2073.
19. Haggstrom, M., Medical gallery of Mikael Haggstrom 2014. Wikiversity Journal of Medicine, 2014. 1(2).
20. Bindu A, H. and S. B, Potency of Various Types of Stem Cells and their Transplantation. Journal of Stem Cell Research & Therapy, 2011. 01(03).
21. Forostyak, O., G. Dayanithi, and S. Forostyak, CNS regenerative medicine and stem cells. Opera Medica et Physiologica, 2016(1).
22. Thomson, J.A., et al., Embryonic stem cell lines derived from human blastocysts. science, 1998. 282(5391): p. 1145-1147.
23. Yabut, O. and H.S. Bernstein, The promise of human embryonic stem cells in aging-associated diseases. Aging (Albany NY), 2011. 3(5): p. 494.
24. 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. 663-676.
25. Takahashi, K., et al., Induction of pluripotent stem cells from adult human fibroblasts by defined factors. cell, 2007. 131(5): p. 861-872.
26. Yu, J., et al., Induced pluripotent stem cell lines derived from human somatic cells. Science, 2007. 318(5858): p. 1917-1920.
27. Narsinh, K.H., J. Plews, and J.C. Wu, Comparison of human induced pluripotent and embryonic stem cells: fraternal or identical twins? Molecular Therapy, 2011. 19(4): p. 635-638.
28. Kropp, C., D. Massai, and R. Zweigerdt, Progress and challenges in large-scale expansion of human pluripotent stem cells. Process Biochemistry, 2017. 59: p. 244-254.
29. Jamshaid, T., et al., Magnetic particles: From preparation to lab-on-a-chip, biosensors, microsystems and microfluidics applications. TrAC Trends in Analytical Chemistry, 2016. 79: p. 344-362.
30. Bilousova, G. and D.R. Roop, Induced Pluripotent Stem Cells in Dermatology: Potentials, Advances, and Limitations. Cold Spring Harbor Perspectives in Medicine, 2014. 4(11): p. 15.
31. 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.
32. 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.
33. 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. e10246.
34. Hwang, S.-T., et al., The expansion of human ES and iPS cells on porous membranes and proliferating human adipose-derived feeder cells. Biomaterials, 2010. 31(31): p. 8012-8021.
35. Hughes, C.S., L.M. Postovit, and G.A. Lajoie, Matrigel: a complex protein mixture required for optimal growth of cell culture. Proteomics, 2010. 10(9): p. 1886-1890.
36. Wang, H., X. Luo, and J. Leighton, Extracellular matrix and integrins in embryonic stem cell differentiation. Biochemistry insights, 2015. 8: p. BCI. S30377.
37. Higuchi, A., et al., Biomimetic cell culture proteins as extracellular matrices for stem cell differentiation. Chemical reviews, 2012. 112(8): p. 4507-4540.
38. Hynes, R.O., Integrins: versatility, modulation, and signaling in cell adhesion. Cell, 1992. 69(1): p. 11-25.
39. 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.
40. Melkoumian, Z., et al., Synthetic peptide-acrylate surfaces for long-term self-renewal and cardiomyocyte differentiation of human embryonic stem cells. Nat Biotechnol, 2010. 28(6): p. 606-10.
41. 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.
42. 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.
43. Kolhar, P., et al., Synthetic surfaces for human embryonic stem cell culture. J Biotechnol, 2010. 146(3): p. 143-6.
44. Klim, J.R., et al., A defined glycosaminoglycan-binding substratum for human pluripotent stem cells. Nature methods, 2010. 7(12): p. 989.
45. 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.
46. 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.
47. Banks, J.M., et al., The combined effects of matrix stiffness and growth factor immobilization on the bioactivity and differentiation capabilities of adipose-derived stem cells. Biomaterials, 2014. 35(32): p. 8951-8959.
48. 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.
49. Engler, A.J., et al., Matrix elasticity directs stem cell lineage specification. Cell, 2006. 126(4): p. 677-689.
50. 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.
51. 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.
52. 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.
53. 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.
54. 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.
55. 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.
56. 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.
57. 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.
58. Bai, S., et al., Silk scaffolds with tunable mechanical capability for cell differentiation. Acta biomaterialia, 2015. 20: p. 22-31.
59. 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.
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, 2017. 37(7): p. 647-660.
61. Ye, K., et al., Matrix stiffness and nanoscale spatial organization of cell-adhesive ligands direct stem cell fate. Nano letters, 2015. 15(7): p. 4720-4729.
62. Wen, J.H., et al., Interplay of matrix stiffness and protein tethering in stem cell differentiation. Nature materials, 2014. 13(10): p. 979.
63. Goetzke, R., et al., Does soft really matter? Differentiation of induced pluripotent stem cells into mesenchymal stromal cells is not influenced by soft hydrogels. Biomaterials, 2018. 156: p. 147-158.
64. 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.
65. 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.
66. Mei, Y., et al., Combinatorial development of biomaterials for clonal growth of human pluripotent stem cells. Nature materials, 2010. 9(9): p. 768.
67. Higuchi, A., et al., Growth of L929 cells on polymeric films prepared by Langmuir–Blodgett and casting methods. Journal of Biomaterials Science, Polymer Edition, 2000. 11(2): p. 149-168.
68. Ullmann, U., et al., Epithelial–mesenchymal transition process in human embryonic stem cells cultured in feeder-free conditions. Molecular human reproduction, 2006. 13(1): p. 21-32.
69. Dedhar, S., Cell–substrate interactions and signaling through ILK. Current opinion in cell biology, 2000. 12(2): p. 250-256.
70. 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(1): p. 60.
71. Pashuck, E.T. and M.M. Stevens, Designing regenerative biomaterial therapies for the clinic. Science translational medicine, 2012. 4(160): p. 160sr4-160sr4.
72. Armstrong, L., et al., The role of PI3K/AKT, MAPK/ERK and NFκβ signalling in the maintenance of human embryonic stem cell pluripotency and viability highlighted by transcriptional profiling and functional analysis. Human molecular genetics, 2006. 15(11): p. 1894-1913.
73. Llewelyn, C., et al., Possible transmission of variant Creutzfeldt-Jakob disease by blood transfusion. The Lancet, 2004. 363(9407): p. 417-421.
74. Ludwig, T. and J. A Thomson, Defined, feeder?independent medium for human embryonic stem cell culture. Current protocols in stem cell biology, 2007: p. 1C. 2.1-1C. 2.16.
75. Ludwig, T.E., et al., Derivation of human embryonic stem cells in defined conditions. Nature biotechnology, 2006. 24(2): p. 185.
76. Chen, G., et al., Chemically defined conditions for human iPSC derivation and culture. Nature methods, 2011. 8(5): p. 424.
77. Furue, M.K., et al., Heparin promotes the growth of human embryonic stem cells in a defined serum-free medium. Proceedings of the National Academy of Sciences, 2008. 105(36): p. 13409-13414.
78. Li, Y., et al., Expansion of human embryonic stem cells in defined serum?free medium devoid of animal?derived products. Biotechnology and bioengineering, 2005. 91(6): p. 688-698.
79. Desai, N., P. Rambhia, and A. Gishto, Human embryonic stem cell cultivation: historical perspective and evolution of xeno-free culture systems. Reproductive Biology and Endocrinology, 2015. 13(1): p. 9.
80. Levenstein, M.E., et al., Basic fibroblast growth factor support of human embryonic stem cell self?renewal. Stem cells, 2006. 24(3): p. 568-574.
81. Vallier, L., M. Alexander, and R.A. Pedersen, Activin/Nodal and FGF pathways cooperate to maintain pluripotency of human embryonic stem cells. Journal of cell science, 2005. 118(19): p. 4495-4509.
82. James, D., et al., TGFβ/activin/nodal signaling is necessary for the maintenance of pluripotency in human embryonic stem cells. Development, 2005. 132(6): p. 1273-1282.
83. Vallier, L., et al., Activin/Nodal signalling maintains pluripotency by controlling Nanog expression. Development, 2009. 136(8): p. 1339-1349.
84. Vallier, L., D. Reynolds, and R.A. Pedersen, Nodal inhibits differentiation of human embryonic stem cells along the neuroectodermal default pathway. Developmental biology, 2004. 275(2): p. 403-421.
85. Pellegrini, L., et al., Crystal structure of fibroblast growth factor receptor ectodomain bound to ligand and heparin. Nature, 2000. 407(6807): p. 1029.
86. Roghani, M., et al., Heparin increases the affinity of basic fibroblast growth factor for its receptor but is not required for binding. Journal of Biological Chemistry, 1994. 269(6): p. 3976-3984.
87. Pantoliano, M.W., et al., Multivalent ligand-receptor binding interactions in the fibroblast growth factor system produce a cooperative growth factor and heparin mechanism for receptor dimerization. Biochemistry, 1994. 33(34): p. 10229-10248.
88. Pauklin, S. and L. Vallier, Activin/Nodal signalling in stem cells. Development, 2015. 142(4): p. 607-619.
89. Sussman, E.M., M.B. Clarke, and V.P. Shastri, Single-step process to produce surface-functionalized polymeric nanoparticles. Langmuir, 2007. 23(24): p. 12275-12279.
90. Gospodarowicz, D., et al., Structural characterization and biological functions of fibroblast growth factor. Endocrine reviews, 1987. 8(2): p. 95-114.
91. Geiger, M., R. Li, and W. Friess, Collagen sponges for bone regeneration with rhBMP-2. Advanced drug delivery reviews, 2003. 55(12): p. 1613-1629.
92. Santo, V.t.E.r., et al., Carrageenan-based hydrogels for the controlled delivery of PDGF-BB in bone tissue engineering applications. Biomacromolecules, 2009. 10(6): p. 1392-1401.
93. Park, H., et al., Injectable biodegradable hydrogel composites for rabbit marrow mesenchymal stem cell and growth factor delivery for cartilage tissue engineering. Biomaterials, 2007. 28(21): p. 3217-3227.
94. Kuo, Y.-C. and M.-J. Huang, Material-driven differentiation of induced pluripotent stem cells in neuron growth factor-grafted poly (ε-caprolactone)-poly (β-hydroxybutyrate) scaffolds. Biomaterials, 2012. 33(23): p. 5672-5682.
95. Ma, F., et al., Neural stem/progenitor cells on collagen with anchored basic fibroblast growth factor as potential natural nerve conduits for facial nerve regeneration. Acta biomaterialia, 2017. 50: p. 188-197.
96. Herland, A., et al., Electrochemical Control of Growth Factor Presentation To Steer Neural Stem Cell Differentiation. Angewandte Chemie-International Edition, 2011. 50(52): p. 12529-12533.
97. Braghirolli, D.I., et al., Electrospun scaffolds functionalized with heparin and vascular endothelial growth factor increase the proliferation of endothelial progenitor cells. Biomedical Materials, 2017. 12(2): p. 16.
98. Benoit, D.S. and K.S. Anseth, Heparin functionalized PEG gels that modulate protein adsorption for hMSC adhesion and differentiation. Acta biomaterialia, 2005. 1(4): p. 461-470.
99. Ayerst, B.I., et al., Growth Differentiation Factor 5-Mediated Enhancement of Chondrocyte Phenotype Is Inhibited by Heparin: Implications for the Use of Heparin in the Clinic and in Tissue Engineering Applications. Tissue Engineering Part A, 2017. 23(7-8): p. 275-292.
100. Wang, Z.G., et al., Fibroblast Growth Factor-1 Released from a Heparin Coacervate Improves Cardiac Function in a Mouse Myocardial Infarction Model. Acs Biomaterials Science & Engineering, 2017. 3(9): p. 1988-1999.
101. Steffens, G., et al., Modulation of angiogenic potential of collagen matrices by covalent incorporation of heparin and loading with vascular endothelial growth factor. Tissue engineering, 2004. 10(9-10): p. 1502-1509.
102. Yoon, J.J., et al., Heparin?immobilized biodegradable scaffolds for local and sustained release of angiogenic growth factor. Journal of Biomedical Materials Research Part A, 2006. 79(4): p. 934-942.
103. Smith Jr, R.J., et al., Capture of endothelial cells under flow using immobilized vascular endothelial growth factor. Biomaterials, 2015. 51: p. 303-312.
104. Landesberg, R., M. Roy, and R.S. Glickman, Quantification of growth factor levels using a simplified method of platelet-rich plasma gel preparation. Journal of Oral and Maxillofacial Surgery, 2000. 58(3): p. 297-300.
105. Eppley, B.L., J.E. Woodell, and J. Higgins, Platelet quantification and growth factor analysis from platelet-rich plasma: implications for wound healing. Plastic and reconstructive surgery, 2004. 114(6): p. 1502-1508.
106. Bredehoft, M., W. Schanzer, and M. Thevis, Quantification of human insulin?like growth factor?1 and qualitative detection of its analogues in plasma using liquid chromatography/electrospray ionisation tandem mass spectrometry. Rapid Communications in Mass Spectrometry, 2008. 22(4): p. 477-485.
107. ?tefkova, K., J. Prochazkova, and J. Pachernik, Alkaline phosphatase in stem cells. Stem cells international, 2015. 2015.
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. 1109-1116.
109. Masui, S., et al., Pluripotency governed by Sox2 via regulation of Oct3/4 expression in mouse embryonic stem cells. Nature cell biology, 2007. 9(6): p. 625.
110. Xu, N., et al., MicroRNA-145 regulates OCT4, SOX2, and KLF4 and represses pluripotency in human embryonic stem cells. Cell, 2009. 137(4): p. 647-658.
111. Itskovitz-Eldor, J., et al., Differentiation of human embryonic stem cells into embryoid bodies compromising the three embryonic germ layers. Molecular medicine, 2000. 6(2): p. 88.
112. Kurosawa, H., Methods for inducing embryoid body formation: in vitro differentiation system of embryonic stem cells. Journal of bioscience and bioengineering, 2007. 103(5): p. 389-398.
113. Sheridan, S.D., V. Surampudi, and R.R. Rao, Analysis of embryoid bodies derived from human induced pluripotent stem cells as a means to assess pluripotency. Stem cells international, 2012. 2012.
114. Reubinoff, B.E., et al., Embryonic stem cell lines from human blastocysts: somatic differentiation in vitro. Nature biotechnology, 2000. 18(4): p. 399.
115. Fishbein, I., et al., Adenoviral vector tethering to metal surfaces via hydrolyzable cross-linkers for the modulation of vector release and transduction. Biomaterials, 2013. 34(28): p. 6938-6948.
116. Finley, M.J., et al., Diminished adhesion and activation of platelets and neutrophils with CD47 functionalized blood contacting surfaces. Biomaterials, 2012. 33(24): p. 5803-5811.
117. Movassaghian, S., et al., Post-Transcriptional Regulation of the GASC1 Oncogene with Active Tumor-Targeted siRNA-Nanoparticles. Molecular pharmaceutics, 2016. 13(8): p. 2605-2621.
118. Boucher, C., et al., Human corneal epithelial cell response to epidermal growth factor tethered via coiled-coil interactions. Biomaterials, 2010. 31(27): p. 7021-7031.
119. 李孟蓓, 人類胚胎幹細胞在無滋養層及無異種條件的培養下於帶有生長因子的表面進行培養; Human Embryonic Stem Cells Culture on Growth Factor-immobilized Surface under Feeder-free and Xeno-free Conditions. 2017, 國立中央大學.
指導教授 劉青原(Ching-Yuan Liu) 審核日期 2018-8-13
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