博碩士論文 106324035 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:22 、訪客IP:3.145.50.196
姓名 江逸蓬(Yi-Peng Jiang)  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 藉由分離及培養初代羊水幹細胞取得並分析通用誘導多能幹細胞的特性
(Generation and characterization of universal induced pluripotent stem cells by isolation of primary amniotic fluid stem cells)
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]   至系統瀏覽論文 (2025-7-23以後開放)
摘要(中) 每年數百萬人由於事故、先天缺陷和疾病致使器官和組織的損失及傷害。對於細胞治療而言,幹細胞是相當吸引人的資源。多能幹細胞,如人類胚胎幹細胞(hESCs)和人類誘導多能幹細胞(hiPSCs),具有分化為三個胚層中任何細胞類型的潛力。如今,由於人類誘導多能幹細胞其多能性、無限增殖且沒有倫理問題,已為治療學的研究增加許多可能。人類誘導多能幹細胞(hiPSCs)可以透過成體幹細胞引入轉錄因子(例如Oct3/4, Sox2, c-myc, klf-4)進行重新編程而得。然而過去必須建立特定患者的細胞株或是人類白血球抗原(HLA)的幹細胞庫以避免治療產生免疫反應,這是耗時耗力且昂貴的。在這項研究中,我們成功自人類羊水幹細胞(hAFSCs)中取得通用人類誘導多能幹細胞,即使將此細胞分化為特定譜系,也不會顯示第一類(HLA-A, -B, and -C)和第二類人類白血球抗原(HLA)。此通用人類誘導多能幹細胞將成為幹細胞治療最有淺力的細胞來源。首先,我們利用以下不同的培養基分離初代的人類羊水幹細胞(1)20%胎牛血清(FBS)培養基(2)20%胎牛血清與等體積羊水混合(FBS+AF)之培養基(3)10%人類血小板裂解液(hPL)培養基(4)羊水(AF)。使用20%胎牛血清與等體積羊水混合(FBS+AF)之培養基可以使初代羊水幹細胞貼附最多並成長最快。因此利用此方式分離並建立後續實驗所需之細胞株。我們假設可從某些細胞系(例如胎兒細胞(hAFSCs))取得並產生通用人類誘導多能幹細胞。在室溫下混合兩個至是五個不同個體捐贈之羊水兩天後,置於細胞培養皿(TCPS)中。不同個體的羊水成分以及單核球在混合期間將產生免疫反應,僅有通用人類羊水幹細胞存活。我們成功的利用仙台病毒轉染多能基因(山中伸彌因子)將(a)混合的人類羊水幹細胞以及(b)單一的人類羊水幹細胞轉為人類誘導多能幹細胞。藉由流式細胞儀檢測第一類(HLA-A, -B, and -C)及第二類人類白血球抗原於通用人類誘導多能幹細胞及其分化之心肌細胞之表現,結果為陰性。即使利用不同捐贈者取得之單核球處理通用人類誘導多能幹細胞分化之心肌細胞,此心肌細胞仍可持續跳動。因此,理論上我們取得之通用人類誘導多能幹細胞僅需單一細胞株便可用於所有不同的患者進行幹細胞治療。
摘要(英) Millions of people suffer from loss and damage of organs and tissue every year due to accidents, birth defects and disease. Stem cells are an attractive source for cell therapy. Pluripotent stem cells such as human embryonic stem cells (hESCs) and human induced pluripotent stem cells (hiPSCs) have the potential to differentiate into any types of the cells derived from all three germ layers. Nowadays, hiPSCs have opened up many gateways for the research in therapeutics because of their pluripotency and unlimited proliferation with no ethical concern. hiPSCs can be reprogrammed from adult somatic cells by introducing transcription factors such as Oct3/4, Sox2, c-myc and klf-4. However, it is necessary to establish patient-specific stem cell line or stem cells bank having specific human leukocyte antigen (HLA) to avoid immune response of the patients. The stem cell bank is laborious, time-consuming and extremely expensive. In this study, universal hiPSCs derived from human amniotic fluid stem cells (AFSCs) have been developed; these cells do not or less express human leukocyte antigen (HLA) class Ia (HLA-A, -B, and -C) and class II even after differentiation of universal hiPSCs into specific lineages of the cells. These universal hiPSCs will be a promising source for stem cell therapy. At first, primary AFSCs were isolated using different media: (1) 20% fetal bovine serum (FBS) medium, (2) 20% FBS medium mixed with equal volume of amniotic fluid (FBS+AF), (3) 10% human platelet lysate (hPL) and (4) amniotic fluid (AF). The isolation of primary AFSCs using FBS+AF medium showed the highest attachment and proliferation of AFSCs. Therefore, this isolation protocol was used to establish hAFSCs in this study. It is hypothesized that hiPSCs derived from some cell lines such as fetal cells, e.g., hAFSCs, may be generated as universal hiPSCs. AFs from two or five donors were mixed and incubated in room temperature for two days and subsequently seeded on conventional tissue culture polystyrene (TCPS) plate. During mixing AFs from different donors, mononuclear cells or components of AFs in different person are supposed to generate immunogenic reaction, which targeted to different hAFSCs from each other and only “universal hAFSCs” are survived after mixing more than two donor of AFs. We succeeded to generate hiPSCs using (a) hAFSCs derived mixed AF [hiPSCs (mix)] and (b) hAFSCs from a single AF [hiPSCs (single)], where these hiPSCs were generated from transfection of pluripotent genes (Yamanaka factors) using Sendai virus vector. HLA Class Ia (HLA-A, -B, and -C) and Class II of universal hiPSCs (mix) and cardiomyocytes differentiated from universal hiPSCs (mix) were evaluated using flow cytometry. The results showed that the universal hiPSCs (mix) did not express HLA Class Ia (HLA-A, -B, and -C) and Class II even after differentiation into cardiomyocytes. Cardiomyocytes differentiated from universal hiPSCs (mix) survived and continued beating even after treatment with mononuclear cells derived from different donors. Universal hiPSCs (mix) should be suitable for stem cell therapy, because universal hiPSCs can theoretically be used to treat any patients using only one cell line.
關鍵字(中) ★ 通用誘導多能幹細胞
★ 幹細胞
★ 誘導多能幹細胞
關鍵字(英)
論文目次 Abstract i
摘要…………………………………………………………………………………………… ii
Index of Content iii
Index of Figure v
Index of Table xi
Chapter 1 Introduction 1
1-1 Stem Cell Application in Regenerative Medicine 1
1-2 Stem Cells 3
1-2-1 Human Pluripotent Stem Cells (hPSCs) 3
1-2-2 Human Mesenchymal Stem Cells (hMSCs) 7
1-3 Human Amniotic Fluid Stem Cells (hAFSCs) 8
1-3-1 Source of Human Amniotic Fluid Stem Cells 10
1-4 Extracellular Matrix (ECM) 13
1-5 Peripheral Blood Mononuclear Cells 14
1-6 Recent developments of universal hiPSCs 15
1-7 Goal of this research 17
Chapter 2 Materials and methods 19
2-1 Experimental materials 19
2-1-1 Cell source for cultivation 19
2-1-2 ECM-coated dish for cell culture 22
2-1-3 Differentiation medium 22
2-1-4 Characterization of stem cells 24
2-2 Experimental instruments 27
2-3 Experimental methods 27
2-3-1 Preparation of the cell culture medium and buffer solution 28
2-3-2 Generation and isolation of stem cell lines 30
2-3-3 Cultivation and passage of stem cells 34
2-3-4 Cell number and seeding density 36
2-3-5 Isolation of mononuclear cells 38
2-3-6 Live and Dead staining 39
2-3-7 Cardiomyocytes differentiation 40
2-3-8 Immunofluorescence staining 41
2-3-9 Flow cytometry measurements 43
2-3-10 Embryoid Body (EB) Formation in Vitro 43
2-3-11 Teratoma Formation in Vivo 44
Chapter 3 Results and Discussion 46
3-1 HLA class I and class II expression of several human stem cells and cancer cells 46
3-2 Cultivation of hAFSCs 47
3-2-1 The morphology of primary hAFSCs isolated using different culture medium 47
3-2-2 The morphology of single and mixing hAFSCs 50
3-2-3 Identification of the hAFSCs from surface marker analysis using flow cytometry 53
3-2-4 The morphology of hAFSCs on ECMs-coating dishes 56
3-3 Reprogramming and cultivation of hAFSCs-derived hiPSCs 58
3-3-1 Generation and isolation of hAFSCs-derived hiPSCs 59
3-3-2 Immunostaining analysis of hAFSCs-derived hiPSCs 62
3-4 Cardiomyocytes (CMs) differentiation 62
3-4-1 The morphology of hiPSCs-derived CMs 63
3-4-2 Immunostaining analysis of hiPSCs-derived CMs 65
3-5 HLA class I and class II expression and evaluation of the immunogenic reaction using mononuclear cells 65
3-5-1 HLA class I and class II expression of hESCs, hiPSCs, hAFSCs, and universal hiPSCs 66
3-5-2 Evaluation of the immunogenic reaction using mononuclear cells 69
3-6 Differentiation ability in vitro 71
3-7 Differentiation ability in vivo 72
Chapter 4 Conclusion 73
Reference 74
Supplementary Data 81
參考文獻 1. Galiniak, S., et al., “The possibilities of stem cell application in regenerative medicine”. Polish Hyperbaric Research, 2016. 54 (1): p. 49.
2. Samsonraj, R.M., et al., “Concise Review: Multifaceted Characterization of Human Mesenchymal Stem Cells for Use in Regenerative Medicine”. STEM CELLS Translational Medicine, 2017. 6 (12): p. 2173.
3. Biehl, J.K. and B. Russell, “Introduction to stem cell therapy”. The Journal of cardiovascular nursing, 2009. 24 (2): p. 98.
4. Marks, P. and S. Gottlieb, “Balancing safety and innovation for cell-based regenerative medicine”. New England Journal of Medicine, 2018. 378 (10): p. 954.
5. Mahla, R.S., “Stem Cells Applications in Regenerative Medicine and Disease Therapeutics”. International Journal of Cell Biology, 2016. 2016: p. 6940283.
6. Karussis, D., et al., “Safety and immunological effects of mesenchymal stem cell transplantation in patients with multiple sclerosis and amyotrophic lateral sclerosis”. Archives of neurology, 2010. 67 (10): p. 1187.
7. Yubo, M., et al., “Clinical efficacy and safety of mesenchymal stem cell transplantation for osteoarthritis treatment: a meta-analysis”. PloS one, 2017. 12 (4).
8. Lee, H.C., et al., “Safety and effect of adipose tissue-derived stem cell implantation in patients with critical limb ischemia”. Circulation Journal, 2012: p. 1204091686.
9. Kawamura, M., et al., “Feasibility, safety, and therapeutic efficacy of human induced pluripotent stem cell-derived cardiomyocyte sheets in a porcine ischemic cardiomyopathy model”. Circulation, 2012. 126 (11_suppl_1): p. S29.
10. Zietlow, R., et al., “Human stem cells for CNS repair”. Cell and tissue research, 2008. 331 (1): p. 301.
11. Yi, S., et al., “Extracellular matrix scaffolds for tissue engineering and regenerative medicine”. Current stem cell research & therapy, 2017. 12 (3): p. 233.
12. Masumoto, H., et al., “Human iPS cell-engineered cardiac tissue sheets with cardiomyocytes and vascular cells for cardiac regeneration”. Scientific reports, 2014. 4: p. 6716.
13. Patel, N.G. and G. Zhang, “Responsive systems for cell sheet detachment”. Organogenesis, 2013. 9 (2): p. 93.
14. Choi, A., et al., “Bulk poly (N-isopropylacrylamide)(PNIPAAm) thermoresponsive cell culture platform: toward a new horizon in cell sheet engineering”. Biomaterials science, 2019. 7 (6): p. 2277.
15. Sung, T.-C., et al., “Efficient differentiation of human pluripotent stem cells into cardiomyocytes on cell sorting thermoresponsive surface”. Biomaterials, 2020: p. 120060.
16. Matsuura, K., et al., “Cell sheet transplantation for heart tissue repair”. Journal of controlled release, 2013. 169 (3): p. 336.
17. Lane, S.W., et al., “Modulating the stem cell niche for tissue regeneration”. Nature biotechnology, 2014. 32 (8): p. 795.
18. Frese, L., et al., “Adipose tissue-derived stem cells in regenerative medicine”. Transfusion Medicine and Hemotherapy, 2016. 43 (4): p. 268.
19. Squillaro, T., et al., “Clinical trials with mesenchymal stem cells: an update”. Cell transplantation, 2016. 25 (5): p. 829.
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. Kaebisch, C., et al., “The role of purinergic receptors in stem cell differentiation”. Computational and structural biotechnology journal, 2015. 13: p. 75.
22. Takahashi, K., et al., “Induction of pluripotent stem cells from adult human fibroblasts by defined factors”. Cell, 2007. 131 (5): p. 861.
23. Hu, B.-Y., et al., “Neural differentiation of human induced pluripotent stem cells follows developmental principles but with variable potency”. Proceedings of the National Academy of Sciences, 2010. 107 (9): p. 4335.
24. Lian, X., et al., “Robust cardiomyocyte differentiation from human pluripotent stem cells via temporal modulation of canonical Wnt signaling”. Proceedings of the National Academy of Sciences, 2012. 109 (27): p. E1848.
25. Thomas, R.J., et al., “Automated, scalable culture of human embryonic stem cells in feeder‐free conditions”. Biotechnology and Bioengineering, 2009. 102 (6): p. 1636.
26. Braam, S.R., et al., “Cardiomyocytes from human pluripotent stem cells in regenerative medicine and drug discovery”. Trends in pharmacological sciences, 2009. 30 (10): p. 536.
27. Nostro, M.C. and G. Keller. Generation of beta cells from human pluripotent stem cells: potential for regenerative medicine. in Seminars in cell & developmental biology. 2012. Elsevier.
28. Tabar, V. and L. Studer, “Pluripotent stem cells in regenerative medicine: challenges and recent progress”. Nature Reviews Genetics, 2014. 15 (2): p. 82.
29. Thomson, J.A., et al., “Embryonic stem cell lines derived from human blastocysts”. science, 1998. 282 (5391): p. 1145.
30. Pera, M.F., et al., “Human embryonic stem cells”. J Cell Sci, 2000. 113 (1): p. 5.
31. Stojkovic, M., et al., “Derivation of human embryonic stem cells from day‐8 blastocysts recovered after three‐step in vitro culture”. Stem Cells, 2004. 22 (5): p. 790.
32. Mitalipova, M., et al., “Human embryonic stem cell lines derived from discarded embryos”. Stem Cells, 2003. 21 (5): p. 521.
33. Vazin, T. and W.J. Freed, “Human embryonic stem cells: derivation, culture, and differentiation: a review”. Restorative Neurology and Neuroscience, 2010. 28 (4): p. 589.
34. Landry, D.W. and H.A. Zucker, “Embryonic death and the creation of human embryonic stem cells”. The Journal of clinical investigation, 2004. 114 (9): p. 1184.
35. Robertson, J.A., “Human embryonic stem cell research: ethical and legal issues”. Nature Reviews Genetics, 2001. 2 (1): p. 74.
36. Wert, G.d. and C. Mummery, “Human embryonic stem cells: research, ethics and policy”. Human reproduction, 2003. 18 (4): p. 672.
37. Okita, K., et al., “Generation of germline-competent induced pluripotent stem cells”. Nature, 2007. 448 (7151): p. 313.
38. 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.
39. Yu, J., et al., “Induced pluripotent stem cell lines derived from human somatic cells”. science, 2007. 318 (5858): p. 1917.
40. Stadtfeld, M., et al., “Induced pluripotent stem cells generated without viral integration”. science, 2008. 322 (5903): p. 945.
41. Okita, K., et al., “Generation of mouse induced pluripotent stem cells without viral vectors”. science, 2008. 322 (5903): p. 949.
42. Okita, K., et al., “A more efficient method to generate integration-free human iPS cells”. Nature methods, 2011. 8 (5): p. 409.
43. Kaji, K., et al., “Virus-free induction of pluripotency and subsequent excision of reprogramming factors”. Nature, 2009. 458 (7239): p. 771.
44. Woltjen, K., et al., “piggyBac transposition reprograms fibroblasts to induced pluripotent stem cells”. Nature, 2009. 458 (7239): p. 766.
45. Yusa, K., et al., “Generation of transgene-free induced pluripotent mouse stem cells by the piggyBac transposon”. Nature methods, 2009. 6 (5): p. 363.
46. Warren, L., et al., “Highly efficient reprogramming to pluripotency and directed differentiation of human cells with synthetic modified mRNA”. Cell Stem Cell, 2010. 7 (5): p. 618.
47. 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.
48. Kim, D., et al., “Generation of human induced pluripotent stem cells by direct delivery of reprogramming proteins”. Cell Stem Cell, 2009. 4 (6): p. 472.
49. Zhou, H., et al., “Generation of induced pluripotent stem cells using recombinant proteins”. Cell Stem Cell, 2009. 4 (5): p. 381.
50. Kim, Y., et al., “Small molecule-mediated reprogramming of human hepatocytes into bipotent progenitor cells”. Journal of hepatology, 2019. 70 (1): p. 97.
51. Fusaki, N., et al., “Efficient induction of transgene-free human pluripotent stem cells using a vector based on Sendai virus, an RNA virus that does not integrate into the host genome”. Proceedings of the Japan Academy, Series B, 2009. 85 (8): p. 348.
52. Seki, T., et al., “Generation of induced pluripotent stem cells from human terminally differentiated circulating T cells”. Cell Stem Cell, 2010. 7 (1): p. 11.
53. Huang, C.Y., et al., “Human iPSC banking: barriers and opportunities”. Journal of Biomedical Science, 2019. 26 (1): p. 87.
54. Levy, D., et al., “Oxysterols and mesenchymal stem cell biology”. Chemistry and physics of lipids, 2017. 207: p. 223.
55. Dominici, M., et al., “Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement”. Cytotherapy, 2006. 8 (4): p. 315.
56. Caplan, A.I., “Mesenchymal stem cells”. Journal of orthopaedic research, 1991. 9 (5): p. 641.
57. Volarevic, V., et al., “Ethical and safety issues of stem cell-based therapy”. International journal of medical sciences, 2018. 15 (1): p. 36.
58. Yi, T. and S.U. Song, “Immunomodulatory properties of mesenchymal stem cells and their therapeutic applications”. Archives of pharmacal research, 2012. 35 (2): p. 213.
59. Mazini, L., et al., “Regenerative capacity of adipose derived stem cells (ADSCs), comparison with mesenchymal stem cells (MSCs)”. International journal of molecular sciences, 2019. 20 (10): p. 2523.
60. Han, Y., et al., “Mesenchymal stem cells for regenerative medicine”. Cells, 2019. 8 (8): p. 886.
61. Martinelli, D., et al., “A humanized system to expand in vitro amniotic fluid-derived stem cells intended for clinical application”. Cytotherapy, 2016. 18 (3): p. 438.
62. Bertolo, A., et al., “An in vitro expansion score for tissue‐engineering applications with human bone marrow‐derived mesenchymal stem cells”. Journal of tissue engineering and regenerative medicine, 2016. 10 (2): p. 149.
63. De Coppi, P., et al., “Isolation of amniotic stem cell lines with potential for therapy”. Nature biotechnology, 2007. 25 (1): p. 100.
64. Borlongan, C.V., “Amniotic fluid as a source of engraftable stem cells”. Brain circulation, 2017. 3 (3): p. 175.
65. Ramasamy, T.S., et al., “Stem cells derived from amniotic fluid: a potential pluripotent-like cell source for cellular therapy?”. Current stem cell research & therapy, 2018. 13 (4): p. 252.
66. Loukogeorgakis, S.P. and P. De Coppi, “Concise review: amniotic fluid stem cells: the known, the unknown, and potential regenerative medicine applications”. Stem Cells, 2017. 35 (7): p. 1663.
67. Spitzhorn, L.-S., et al., “Isolation and molecular characterization of amniotic fluid-derived mesenchymal stem cells obtained from caesarean sections”. Stem cells international, 2017. 2017.
68. Magatti, M., et al., “The immunomodulatory properties of amniotic cells: The two sides of the coin”. Cell transplantation, 2018. 27 (1): p. 31.
69. Schiavo, A.A., et al., “Endothelial properties of third-trimester amniotic fluid stem cells cultured in hypoxia”. Stem cell research & therapy, 2015. 6 (1): p. 209.
70. Moschidou, D., et al., “Valproic acid confers functional pluripotency to human amniotic fluid stem cells in a transgene-free approach”. Molecular Therapy, 2012. 20 (10): p. 1953.
71. Moschidou, D., et al., “Human mid-trimester amniotic fluid stem cells cultured under embryonic stem cell conditions with valproic acid acquire pluripotent characteristics”. Stem cells and development, 2013. 22 (3): p. 444.
72. Su, C.-F., et al., “Application of amniotic fluid stem cells in repairing sciatic nerve injury in minipigs”. Brain research, 2018. 1678: p. 397.
73. Kumar, V., et al., Robbins and Cotran pathologic basis of disease, professional edition e-book. 2014: elsevier health sciences.
74. Hay, E.D., Cell biology of extracellular matrix. 2013: Springer Science & Business Media.
75. Higuchi, A., et al., “Biomimetic cell culture proteins as extracellular matrices for stem cell differentiation”. Chemical reviews, 2012. 112 (8): p. 4507.
76. Chen, K.G., et al., “Human pluripotent stem cell culture: considerations for maintenance, expansion, and therapeutics”. Cell Stem Cell, 2014. 14 (1): p. 13.
77. Plopper, G., et al., The extracellular matrix and cell adhesion, Cells. 2007, Jones and Bartlett, Sudbury.
78. Higuchi, A., et al., “Stem cell therapies for reversing vision loss”. Trends in biotechnology, 2017. 35 (11): p. 1102.
79. Higuchi, A., et al., “Polymeric design of cell culture materials that guide the differentiation of human pluripotent stem cells”. Progress in Polymer Science, 2017. 65: p. 83.
80. Higuchi, A., et al., “Physical cues of biomaterials guide stem cell differentiation fate”. Chemical reviews, 2013. 113 (5): p. 3297.
81. Wood, K.J., et al., “Understanding stem cell immunogenicity in therapeutic applications”. Trends in immunology, 2016. 37 (1): p. 5.
82. Moon, S.-H., et al., “A system for treating ischemic disease using human embryonic stem cell-derived endothelial cells without direct incorporation”. Biomaterials, 2011. 32 (27): p. 6445.
83. Drukker, M., et al., “Characterization of the expression of MHC proteins in human embryonic stem cells”. Proceedings of the National Academy of Sciences, 2002. 99 (15): p. 9864.
84. Li, L., et al., “Human embryonic stem cells possess immune‐privileged properties”. Stem Cells, 2004. 22 (4): p. 448.
85. 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.
86. Taylor, C.J., et al., “Banking on human embryonic stem cells: estimating the number of donor cell lines needed for HLA matching”. The Lancet, 2005. 366 (9502): p. 2019.
87. Nakajima, F., et al., “Human leukocyte antigen matching estimations in a hypothetical bank of human embryonic stem cell lines in the Japanese population for use in cell transplantation therapy”. Stem Cells, 2007. 25 (4): p. 983.
88. Xu, H., et al., “Targeted disruption of HLA genes via CRISPR-Cas9 generates iPSCs with enhanced immune compatibility”. Cell Stem Cell, 2019. 24 (4): p. 566.
89. Han, X., et al., “Generation of hypoimmunogenic human pluripotent stem cells”. Proceedings of the National Academy of Sciences, 2019. 116 (21): p. 10441.
90. Higuchi, A., et al., “Differentiation ability of amniotic fluid-derived stem cells cultured on extracellular matrix-immobilized surface”. Current Nanoscience, 2011. 7 (6): p. 893.
91. Sung, T.-C., et al., “Effect of cell culture biomaterials for completely xeno-free generation of human induced pluripotent stem cells”. Biomaterials, 2020. 230: p. 119638.
92. 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.
93. 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.
94. Sung, T.-C., et al., “The design of a thermoresponsive surface for the continuous culture of human pluripotent stem cells”. Biomaterials, 2019. 221: p. 119411.
95. 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.
96. Higuchi, A., et al., “Direct ex vivo expansion of hematopoietic stem cells from umbilical cord blood on membranes”. Journal of Membrane Science, 2010. 351 (1-2): p. 104.
97. Moreau, J.L. and H.H. Xu, “Mesenchymal stem cell proliferation and differentiation on an injectable calcium phosphate–chitosan composite scaffold”. Biomaterials, 2009. 30 (14): p. 2675.
98. 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.
99. Shevde, N.K. and A.A. Mael, Techniques in embryoid body formation from human pluripotent stem cells, in Basic Cell Culture Protocols. 2013, Springer. p. 535.
100. 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.
101. Zhang, W.Y., et al., Teratoma formation: A tool for monitoring pluripotency in stem cell research, in StemBook [Internet]. 2012, Harvard Stem Cell Institute.
102. Führmann, T., et al., “Injectable hydrogel promotes early survival of induced pluripotent stem cell-derived oligodendrocytes and attenuates longterm teratoma formation in a spinal cord injury model”. Biomaterials, 2016. 83: p. 23.
103. Gholizadeh-Ghalehaziz, S., et al., “A mini overview of isolation, characterization and application of amniotic fluid stem cells”. International journal of stem cells, 2015. 8 (2): p. 115.
104. Davydova, D., et al., “Cell phenotypes in human amniotic fluid”. Acta Naturae (англоязычная версия), 2009. 1 (2 (2)).
105. Roubelakis, M.G., et al., “Amniotic fluid and amniotic membrane stem cells: marker discovery”. Stem cells international, 2012. 2012.
106. Li, L., et al., “Characteristics of human amniotic fluid mesenchymal stem cells and their tropism to human ovarian cancer”. PloS one, 2015. 10 (4).
107. Mummery, C.L., et al., “Differentiation of human embryonic stem cells and induced pluripotent stem cells to cardiomyocytes: a methods overview”. Circulation research, 2012. 111 (3): p. 344.
108. Yamanaka, S., “Strategies and new developments in the generation of patient-specific pluripotent stem cells”. Cell Stem Cell, 2007. 1 (1): p. 39.
109. Moreau, A., et al., “Induced pluripotent stem-cell-derived cardiomyocytes: Cardiac applications, opportunities, and challenges”. Canadian journal of physiology and pharmacology, 2017. 95 (10): p. 1108.
110. Di Baldassarre, A., et al., “Human-induced pluripotent stem cell technology and cardiomyocyte generation: progress and clinical applications”. Cells, 2018. 7 (6): p. 48.
111. Wesselschmidt, R.L., The teratoma assay: an in vivo assessment of pluripotency, in Human Pluripotent Stem Cells. 2011, Springer. p. 231.
指導教授 樋口亞紺 審核日期 2020-7-23
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