博碩士論文 110324039 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:126 、訪客IP:18.117.77.210
姓名 陳諺泓(Yen-Hung 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 人類多能幹細胞的長期培養和分化在混合寡肽接枝的水凝膠表面
(Hydrogel Surface Grafted with Mixed Oligopeptides for Long-Term Cultivation and Differentiation of Human Pluripotent 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 (2025-8-20以後開放)
摘要(中) 人多能幹細胞(hPSCs)包括人胚胎幹細胞(hESCs)和人誘導多能幹細胞(hiPSCs),具有分化成內胚層、外胚層和中胚層等三個胚層的細胞的能力。 近年含異種的Matrigels被用於培養及分化 hPSC,其中Matrigels由含異種的膠原蛋白 IV、層粘連蛋白-111 等組成。此外,含異種的水凝膠的臨床應用受到限制,因為它們源自動物且化學成分不確定。 根據先前的研究單一種細胞外基質 (ECM) 衍生的寡肽接枝水凝膠常被使用於 hPSC 培養和分化。考量到 Matrigels 中包含的多種成分,我們開發混合肽接枝水凝膠,它可以利用細胞上不同種類的整合素來改善 hPSC 增殖和分化。我們使用N-(3-二甲基氨基丙基)-N′-乙基碳二亞胺鹽酸鹽 (EDC) 和 N-羥基琥珀酰亞胺 (NHS) 化學將幾種類型的層粘連蛋白(KKGCGGKGGPMQKMRGDVFSP)和玻連蛋白衍生(GCGGKGGPQVTRGDVFTMP)的寡肽移植到 PVA-IA 水凝膠上。其中將水凝膠的彈性控制在 25.3 kPa(24 小時交聯時間)。 與單一肽接枝水凝膠相比,人 PSC 可以在混合肽接枝水凝膠上有效增殖。 用低濃度混合肽 (100:100 μg/mL) 製備的水凝膠可以很好地貼附、維持多能性及連續繼代超過10次。 此外,在寡肽的第一個序列上插入正氨基酸(K,賴氨酸)有助於提高寡肽接枝水凝膠的表面接枝密度,即 KKGCGGKGGPMQKMRGDVFSP、KGCGGKGGPQVTRGDVFTMP。 這解釋了肽第一位側鏈上的氨基(KKGCGGKGGPMQKMRGDVFSP)比肽主鏈上的氨基(GCGGKGGPMQKMRGDVFSP)更具反應性。 此外,在寡肽上插入帶正電的賴氨酸 (K) 有助於增強水凝膠的 zeta 電位,此特性有利於細胞的貼附。 我們的含有賴氨酸的混合肽接枝水凝膠有望促進 hPSC 的增殖效率和分化為具有低貼附性的分化細胞,例如神經細胞和視網膜色素上皮細胞。
摘要(英) Human pluripotent stem cells (hPSCs) including human embryonic stem cells (hESCs) and human induced pluripotent stem cells (hiPSCs) have the ability to differentiate into the cells derived from three germ layers, such as endoderm, ectoderm and mesoderm. Xeno-containing Matrigels are typically used to culture and differentiate hPSCs where Matrigels consist of xeno-containing collagen IV, laminin-111, etc. Moreover, clinical application of the xeno-containing hydrogels is limited since they are derived from animal and not chemically defined. Typically, single extracellular matrix protein (ECM)-derived oligopeptide-grafted hydrogels have been used for hPSC culture and differentiation in xeno-free culture conditions. Considering multiple components contained in Matrigels, I designed mixed peptide-grafted hydrogels, which may improve hPSC proliferation and differentiation using different binding sites of hPSCs. I prepared vitronectin-derived peptide (GCGGKGGPQVTRGDVFTMP) and laminin β4-derived peptide (KKGCGGKGGPMQKMRGDVFSP)-grafted poly(vinylalcohol-co-itaconic) hydrogels using N-hydroxysuccinimide (NHS)/1-ethyl-3-(-3-dimethylaminopropyl) carbodiimide (EDC) chemistry where the elasticity of the hydrogels was controlled at 25.3 kPa (24h crosslinking time). Human PSCs could proliferate on the mixed peptide-grafted hydrogels compared to single peptide-grafted hydrogels efficiently. Hydrogels prepared with low concentration of mixed peptides (100:100 μg/mL) could support hPSC adhesion and pluripotency. Furthermore, positive amino acid (K, lysine) insertion on the first sequence of the peptide contributed to the enhancement of surface grafting density of peptide-grafted hydrogels, i.e., KKGCGGKGGPMQKMRGDVFSP and KGCGGKGGPQVTRGDVFTMP. This is explained that the amino group on the side chain of the first position of peptide (KKGCGGKGGPMQKMRGDVFSP) is more reactive than the amino group of the main chain of the peptide (GCGGKGGPMQKMRGDVFSP). Furthermore, insertion of positive lysine (K) on the peptide contributed to enhance zeta potential of the hydrogels, which is favor to adhere hPSCs. Our mixed peptide-grafted hydrogels containing lysine and joint peptide are expected to promote efficient proliferation and differentiation of hPSCs into differentiated cells with low adhesion such as neural cells and retinal pigment epithelium cells.
關鍵字(中) ★ 混和寡肽接枝水凝膠
★ 生醫材料
★ 無異種條件培養
★ 人類多能幹細胞
★ 多能幹細胞分化
關鍵字(英) ★ Mixed oligopeptide grafted hydrogel
★ biomaterials
★ Xeno free condition
★ human pluripotent stem cells
★ cell differentiation
論文目次 Abstract I
摘要 III
Index of content IV
Index of figure VIII
Index of Table XIV
Chapter 1 Introduction 1
1-1 Stem cells 1
1-1-1 Human embryonic stem cells 1
1-1-2 Human induced pluripotent stem cells 3
1-1-3 Characterization of human pluripotent stem cells 3
1-1-4 hPSCs for therapeutic application 5
1-2 Cardiomyocytes 7
1-2-1 Characterization of human cardiomyocytes 7
1-2-2 Efficient methods for cardiomyocytes differentiation 8
1-3 Mesenchymal stem cells 9
1-3-1 Characterization of human mesenchymal stem cells 12
1-3-2 Efficient differentiation methods for mesenchymal stem cells 12
1-4 The biomaterial substrates for stem cells cultivation 13
1-4-1 The feeder cell layers for hPSCs cultivation 13
1-4-2 The maintenance of hPSCs on feeder free and xeno-contained proteins 14
1-4-3 The maintenance of hPSCs on feeder free and xeno-free proteins 15
1-4-4 Development and screening of ECM protein-derived peptides 16
1-4-5 Immobilization method of ECM protein-derived peptides 17
1-4-6 Design of peptides with a joint chain and a dual chain 19
1-4-7 ECM protein-derived peptides for hPSCs adhesion and differentiation 20
1-5 The goal of this study 22
Chapter 2 Materials and Method 24
2-1 Materials 24
2-1-1 Cell lines 24
2-1-2 Commercial culture dishes 24
2-1-3 Commercial coated substrates 24
2-1-4 Medium for hPSCs 24
2-1-5 Medium and chemicals for MSCs differentiated from hPSCs 24
2-1-6 Medium and chemicals for cardiomyocytes differentiated from hPSCs 25
2-1-7 Medium and chemicals for cell passages 25
2-1-8 Phosphate buffer saline solution (PBS) 25
2-1-9 The chemicals of Oligopeptide-grafted Hydrogels 26
2-1-10 Chemicals for immunostaining 30
2-1-11 Chemicals for flow cytometry 31
2-2 Experimental instruments 31
2-3 Experimental methods 33
2-3-1 Human pluripotent stem cells maintenance 33
2-3-2 The passage method of human pluripotent stem cells 33
2-3-3 Expansion fold and differentiation ratio of hPSCs 33
2-3-4 Immunostaining of hPSCs 35
2-3-5 Embryoid body (EB) formation in vitro 38
2-3-6 Flow cytometry measurements 38
2-3-7 Freezing of human pluripotent stem cells 39
2-3-8 Thawing of human pluripotent stem cells 39
2-4 Preparation of oligopeptide-grafted hydrogels 40
2-4-1 Preparation of PVA-IA coated surface 40
2-4-2 Preparation of different oligopeptide-grafted PVA-IA hydrogels 40
2-5 Characterization of oligopeptide-grafted hydrogels 41
2-5-1 X-ray photoelectron spectroscopy (XPS) measurements 41
2-5-2 Zeta potential measurements 42
2-5-3 PrimosCR 45 measurements 43
2-6 Differentiation method of Mesenchymal stem cells 43
2-6-1 The protocol for differentiation of MSCs from hPSCs 43
2-6-2 The passage method for MSCs 44
2-7 Differentiation method of Cardiomyocytes 45
2-7-1 The protocol for differentiation of Cardiomyocytes from hPSCs 45
Chapter 3 Results and discussions 47
3-1 Cultivation of hiPSCs (HPS0077) on different peptide-grafted hydrogels 47
3-1-1 Morphologies of hiPSCs (HPS0077) on hydrogels prepared with different concentration of oligopeptides 48
3-1-2 Expansion fold of hiPSCs (HPS0077) on oligopeptide-grafted hydrogels prepared with different concentration of oligopeptides 53
3-1-3 The morphologies of hiPSCs (HPS0077) on different oligopeptide-grafted hydrogels during long-term cultivation 55
3-1-4 Expansion fold of hiPSCs (HPS0077) on different peptide-grafted hydrogels 59
3-1-5 Pluripotency analysis of hiPSCs (HPS0077) cultured on different oligopeptide-grafted hydrogels 64
3-2 Cultivation of hESCs (WA09) on different oligopeptide-grafted hydrogels 70
3-2-1 Morphologies of hESCs (WA09) on oligopeptide-grafted hydrogels prepared with different concentration of oligopeptides 70
3-2-2 Expansion fold of hESCs (WA09) on oligopeptide-grafted hydrogels prepared with different concentration of oligopeptides 75
3-2-3 Morphologies of hESCs (WA09) on different oligopeptide-grafted hydrogels for Long-term cultivation 77
3-2-4 Expansion fold of hESCs (WA09) on different oligopeptide-grafted hydrogels 81
3-2-5 Pluripotency analysis of hESCs (WA09) on different oligopeptide-grafted hydrogels 82
3-3 Differentiation of human pluripotent stem cells into mesenchymal stem cells on different oligopeptide-grafted hydrogels 88
3-3-1 Differentiation of hPSCs into MSCs on different oligopeptide-grafted hydrogels 88
3-3-2 Surface marker expression of hPSCs derived hMSCs on different oligopeptide-grafted hydrogels 90
3-4 Differentiation of human pluripotent stem cells into cardiomyocytes on different oligopeptide-grafted hydrogels 91
3-4-1 Differentiation of hPSCs into cardiomyocytes on different oligopeptide-grafted hydrogels 91
3-4-2 Specific marker expression of Cardiomyocytes derived from hPSCs cultured on different oligopeptide-grafted hydrogels 94
3-5 Characterization of different oligopeptide-grafted hydrogels 95
3-5-1 X-ray photoelectron spectroscopy analysis of different oligopeptide-grafted hydrogels 95
3-5-2 Zeta potential analysis of the surface electrical potential on different oligopeptide-grafted hydrogels 100
3-5-3 PrimosCR 45 analysis of the surface roughness on different oligopeptide-grafted hydrogels 103
3-6 Correlation between cells expansion fold, concentration, and peptide graft density 106
Chapter 4 Conclusion 108
References 110
參考文獻 1. Menon, S., et al., An overview of direct somatic reprogramming: the ins and outs of iPSCs. International journal of molecular sciences, 2016. 17(1): p. 141.
2. Melton, D., ‘Stemness’: definitions, criteria, and standards, in Essentials of stem cell biology. 2014, Elsevier. p. 7-17.
3. Rajabzadeh, N., E. Fathi, and R. Farahzadi, Stem cell-based regenerative medicine. Stem cell investigation, 2019. 6.
4. Marek J. Łos, A.S., Saeid Ghavami,, Chapter 2 - Stem Cells. Stem Cells and Biomaterials for Regenerative Medicine, 2019: p. Pages 5-16,.
5. Hoffman, L.M. and M.K. Carpenter, Characterization and culture of human embryonic stem cells. Nature biotechnology, 2005. 23(6): p. 699-708.
6. Amit, M., et al., Feeder Layer- and Serum-Free Culture of Human Embryonic Stem Cells1. Biology of Reproduction, 2004. 70(3): p. 837-845.
7. Levenberg, S., et al., Endothelial potential of human embryonic stem cells. Blood, 2007. 110(3): p. 806-814.
8. Ho, P.-J., et al., Current applications of human pluripotent stem cells: possibilities and challenges. Cell transplantation, 2012. 21(5): p. 801-814.
9. Thomson, J.A., et al., Embryonic stem cell lines derived from human blastocysts. science, 1998. 282(5391): p. 1145-1147.
10. Lo, B. and L. Parham, Ethical issues in stem cell research. Endocrine reviews, 2009. 30(3): p. 204-213.
11. Takahashi, K., et al., Induction of pluripotent stem cells from adult human fibroblasts by defined factors. cell, 2007. 131(5): p. 861-872.
12. Nishikawa, S.-i., R.A. Goldstein, and C.R. Nierras, The promise of human induced pluripotent stem cells for research and therapy. Nature reviews Molecular cell biology, 2008. 9(9): p. 725-729.
13. Zhang, J., et al., Functional cardiomyocytes derived from human induced pluripotent stem cells. Circulation research, 2009. 104(4): p. e30-e41.
14. 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.
15. 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.
16. Gokhale, P.J. and P.W. Andrews, Characterization of human pluripotent stem cells. 2013, LWW. p. 1031-1034.
17. Chin, M.H., et al., Induced pluripotent stem cells and embryonic stem cells are distinguished by gene expression signatures. Cell stem cell, 2009. 5(1): p. 111-123.
18. McKnight, C.L., et al., Modelling mitochondrial disease in human pluripotent stem cells: what have we learned? International Journal of Molecular Sciences, 2021. 22(14): p. 7730.
19. Bai, H. and Z. Wang, Directing human embryonic stem cells to generate vascular progenitor cells. Gene therapy, 2008. 15(2): p. 89-95.
20. Lovell-Badge, R., The future for stem cell research. Nature, 2001. 414(6859): p. 88-91.
21. Chang, E.-A., et al., Human induced pluripotent stem cells: clinical significance and applications in neurologic diseases. Journal of Korean Neurosurgical Society, 2019. 62(5): p. 493-501.
22. Bongso, A. and M. Richards, History and perspective of stem cell research. Best practice & research Clinical obstetrics & gynaecology, 2004. 18(6): p. 827-842.
23. Singh, V.K., et al., Induced pluripotent stem cells: applications in regenerative medicine, disease modeling, and drug discovery. Frontiers in cell and developmental biology, 2015. 3: p. 2.
24. Liao, S., et al., Potent immunomodulation and angiogenic effects of mesenchymal stem cells versus cardiomyocytes derived from pluripotent stem cells for treatment of heart failure. Stem cell research & therapy, 2019. 10(1): p. 1-13.
25. Mozaffarian, D., et al., Heart disease and stroke statistics—2015 update: a report from the American Heart Association. circulation, 2015. 131(4): p. e29-e322.
26. Zwi, L., et al., Cardiomyocyte differentiation of human induced pluripotent stem cells. Circulation, 2009. 120(15): p. 1513-1523.
27. Sung, T.-C., et al., Efficient differentiation of human pluripotent stem cells into cardiomyocytes on cell sorting thermoresponsive surface. Biomaterials, 2020. 253: p. 120060.
28. 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.
29. 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. 344-358.
30. Kim, J., P. Sachdev, and K. Sidhu, Alginate microcapsule as a 3D platform for the efficient differentiation of human embryonic stem cells to dopamine neurons. Stem cell research, 2013. 11(3): p. 978-989.
31. Higuchi, A., et al., Biomimetic cell culture proteins as extracellular matrices for stem cell differentiation. Chemical reviews, 2012. 112(8): p. 4507-4540.
32. Li, R.A., et al., Bioengineering an electro-mechanically functional miniature ventricular heart chamber from human pluripotent stem cells. Biomaterials, 2018. 163: p. 116-127.
33. Garreta, E., et al., Myocardial commitment from human pluripotent stem cells: Rapid production of human heart grafts. Biomaterials, 2016. 98: p. 64-78.
34. Jiang, B., et al., Generation of cardiac spheres from primate pluripotent stem cells in a small molecule-based 3D system. Biomaterials, 2015. 65: p. 103-114.
35. Shapira-Schweitzer, K., et al., A photopolymerizable hydrogel for 3-D culture of human embryonic stem cell-derived cardiomyocytes and rat neonatal cardiac cells. Journal of molecular and cellular cardiology, 2009. 46(2): p. 213-224.
36. 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. 83-126.
37. Wang, Q., et al., Functional engineered human cardiac patches prepared from nature′s platform improve heart function after acute myocardial infarction. Biomaterials, 2016. 105: p. 52-65.
38. Kerscher, P., et al., Direct hydrogel encapsulation of pluripotent stem cells enables ontomimetic differentiation and growth of engineered human heart tissues. Biomaterials, 2016. 83: p. 383-395.
39. 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.
40. Mushahary, D., et al., Isolation, cultivation, and characterization of human mesenchymal stem cells. Cytometry Part A, 2018. 93(1): p. 19-31.
41. Viswanathan, S., et al., Mesenchymal stem versus stromal cells: International Society for Cell & Gene Therapy (ISCT®) Mesenchymal Stromal Cell committee position statement on nomenclature. Cytotherapy, 2019. 21(10): p. 1019-1024.
42. Pig, J., et al., Inflammatory effects of autologous, genetically modified autologous, allogeneic, and xenogeneic mesenchymal stem cells after intra-articular injection in horses. Veterinary and Comparative Orthopaedics and Traumatology, 2013. 26(06): p. 453-460.
43. int Anker, P.S., et al., Amniotic fluid as a novel source of mesenchymal stem cells for therapeutic transplantation. Blood, 2003. 102(4): p. 1548-1549.
44. Muduli, S., et al., Proliferation and osteogenic differentiation of amniotic fluid-derived stem cells. Journal of Materials Chemistry B, 2017. 5(27): p. 5345-5354.
45. Wang, P.-Y., et al., Pluripotency maintenance of amniotic fluid-derived stem cells cultured on biomaterials. Journal of Materials Chemistry B, 2015. 3(18): p. 3858-3869.
46. Huang, G.-J., S. Gronthos, and S. Shi, Critical reviews in oral biology & medicine: mesenchymal stem cells derived from dental tissues vs. those from other sources: their biology and role in regenerative medicine. Journal of Dental Research, 2009. 88(9): p. 792.
47. Ullah, I., R.B. Subbarao, and G.J. Rho, Human mesenchymal stem cells-current trends and future prospective. Bioscience reports, 2015. 35(2).
48. Zhang, L., et al., Two-step generation of mesenchymal stem/stromal cells from human pluripotent stem cells with reinforced efficacy upon osteoarthritis rabbits by HA hydrogel. Cell & Bioscience, 2021. 11: p. 1-17.
49. Liu, T.-M., Application of mesenchymal stem cells derived from human pluripotent stem cells in regenerative medicine. World Journal of Stem Cells, 2021. 13(12): p. 1826.
50. Jiang, B., et al., Concise review: mesenchymal stem cells derived from human pluripotent cells, an unlimited and quality-controllable source for therapeutic applications. Stem cells, 2019. 37(5): p. 572-581.
51. West, M.D., et al., Adult versus pluripotent stem cell-derived mesenchymal stem cells: The need for more precise nomenclature. Current Stem Cell Reports, 2016. 2: p. 299-303.
52. Merimi, M., et al., The therapeutic potential of mesenchymal stromal cells for regenerative medicine: current knowledge and future understandings. Frontiers in Cell and Developmental Biology, 2021. 9: p. 661532.
53. Pittenger, M.F., et al., Multilineage potential of adult human mesenchymal stem cells. science, 1999. 284(5411): p. 143-147.
54. Mamidi, M.K., et al., Comparative cellular and molecular analyses of pooled bone marrow multipotent mesenchymal stromal cells during continuous passaging and after successive cryopreservation. Journal of cellular biochemistry, 2012. 113(10): p. 3153-3164.
55. Otsuru, S., et al., Improved isolation and expansion of bone marrow mesenchymal stromal cells using a novel marrow filter device. Cytotherapy, 2013. 15(2): p. 146-153.
56. Miao, Z., et al., Isolation of mesenchymal stem cells from human placenta: comparison with human bone marrow mesenchymal stem cells. Cell biology international, 2006. 30(9): p. 681-687.
57. La Rocca, G., et al., Isolation and characterization of Oct-4+/HLA-G+ mesenchymal stem cells from human umbilical cord matrix: differentiation potential and detection of new markers. Histochemistry and cell biology, 2009. 131: p. 267-282.
58. Bieback, K., et al., Critical parameters for the isolation of mesenchymal stem cells from umbilical cord blood. Stem cells, 2004. 22(4): p. 625-634.
59. Salehinejad, P., et al., Comparison of different methods for the isolation of mesenchymal stem cells from human umbilical cord Wharton’s jelly. In Vitro Cellular & Developmental Biology-Animal, 2012. 48: p. 75-83.
60. Christodoulou, I., et al., Comparative evaluation of human mesenchymal stem cells of fetal (Wharton′s jelly) and adult (adipose tissue) origin during prolonged in vitro expansion: considerations for cytotherapy. Stem cells international, 2013. 2013.
61. Yoon, J.H., et al., Comparison of explant-derived and enzymatic digestion-derived MSCs and the growth factors from Wharton’s jelly. BioMed research international, 2013. 2013.
62. Pendleton, C., et al., Mesenchymal stem cells derived from adipose tissue vs bone marrow: in vitro comparison of their tropism towards gliomas. PloS one, 2013. 8(3): p. e58198.
63. Baglioni, S., et al., Characterization of human adult stem‐cell populations isolated from visceral and subcutaneous adipose tissue. The FASEB Journal, 2009. 23(10): p. 3494-3505.
64. Wagner, W., et al., Comparative characteristics of mesenchymal stem cells from human bone marrow, adipose tissue, and umbilical cord blood. Experimental hematology, 2005. 33(11): p. 1402-1416.
65. Higuchi, A., et al., Osteoblast differentiation of amniotic fluid-derived stem cells irradiated with visible light. Tissue engineering Part A, 2011. 17(21-22): p. 2593-2602.
66. Tsai, M.S., et al., Isolation of human multipotent mesenchymal stem cells from second‐trimester amniotic fluid using a novel two‐stage culture protocol. Human reproduction, 2004. 19(6): p. 1450-1456.
67. Cai, J., et al., Generation of human induced pluripotent stem cells from umbilical cord matrix and amniotic membrane mesenchymal cells. Journal of Biological Chemistry, 2010. 285(15): p. 11227-11234.
68. Huang, G.-J., S. Gronthos, and S. Shi, Mesenchymal stem cells derived from dental tissues vs. those from other sources: their biology and role in regenerative medicine. Journal of dental research, 2009. 88(9): p. 792-806.
69. Hilkens, P., et al., Effect of isolation methodology on stem cell properties and multilineage differentiation potential of human dental pulp stem cells. Cell and tissue research, 2013. 353(1): p. 65-78.
70. Seifrtová, M., et al., The response of human ectomesenchymal dental pulp stem cells to cisplatin treatment. International endodontic journal, 2012. 45(5): p. 401-412.
71. Riekstina, U., et al., Characterization of human skin-derived mesenchymal stem cell proliferation rate in different growth conditions. Cytotechnology, 2008. 58: p. 153-162.
72. Bartsch Jr, G., et al., Propagation, expansion, and multilineage differentiation of human somatic stem cells from dermal progenitors. Stem cells and development, 2005. 14(3): p. 337-348.
73. Raynaud, C., et al., Comprehensive characterization of mesenchymal stem cells from human placenta and fetal membrane and their response to osteoactivin stimulation. Stem cells international, 2012. 2012.
74. Rotter, N., et al., Isolation and characterization of adult stem cells from human salivary glands. Stem cells and development, 2008. 17(3): p. 509-518.
75. Sato, A., et al., Isolation, tissue localization, and cellular characterization of progenitors derived from adult human salivary glands. Cloning and stem cells, 2007. 9(2): p. 191-205.
76. Morito, T., et al., Synovial fluid-derived mesenchymal stem cells increase after intra-articular ligament injury in humans. Rheumatology, 2008. 47(8): p. 1137-1143.
77. Hatakeyama, A., et al., Isolation and characterization of synovial mesenchymal stem cell derived from hip joints: a comparative analysis with a matched control knee group. Stem Cells International, 2017. 2017.
78. Heathman, T.R., et al., Characterization of human mesenchymal stem cells from multiple donors and the implications for large scale bioprocess development. Biochemical engineering journal, 2016. 108: p. 14-23.
79. Hynes, K., et al., Generation of functional mesenchymal stem cells from different induced pluripotent stem cell lines. Stem cells and development, 2014. 23(10): p. 1084-1096.
80. Penny, J., et al., The biology of equine mesenchymal stem cells: phenotypic characterization, cell surface markers and multilineage differentiation. Front Biosci, 2012. 17(3): p. 892.
81. Li, E., et al., Generation of mesenchymal stem cells from human embryonic stem cells in a complete serum-free condition. International journal of biological sciences, 2018. 14(13): p. 1901.
82. Higuchi, A., et al., Stem Cell Culture on Polymer Hydrogels. Hydrogels: Recent Advances, 2018: p. 357-408.
83. Polak, U., et al., Selecting and isolating colonies of human induced pluripotent stem cells reprogrammed from adult fibroblasts. JoVE (Journal of Visualized Experiments), 2012(60): p. e3416.
84. Wang, I.-N.E., et al., Apelin enhances directed cardiac differentiation of mouse and human embryonic stem cells. PloS one, 2012. 7(6): p. e38328.
85. Uchida, N., et al., Efficient generation of β-globin-expressing erythroid cells using stromal cell-derived induced pluripotent stem cells from patients with sickle cell disease. Stem Cells, 2017. 35(3): p. 586-596.
86. Llames, S., et al., Feeder layer cell actions and applications. Tissue Engineering Part B: Reviews, 2015. 21(4): p. 345-353.
87. Ghasemi-Dehkordi, P., et al., Comparison between the cultures of human induced pluripotent stem cells (hiPSCs) on feeder-and serum-free system (Matrigel matrix), MEF and HDF feeder cell lines. Journal of cell communication and signaling, 2015. 9: p. 233-246.
88. Aisenbrey, E.A. and W.L. Murphy, Synthetic alternatives to Matrigel. Nature Reviews Materials, 2020. 5(7): p. 539-551.
89. Taub, M., et al., Epidermal growth factor or transforming growth factor alpha is required for kidney tubulogenesis in matrigel cultures in serum-free medium. Proceedings of the National Academy of Sciences, 1990. 87(10): p. 4002-4006.
90. Basic, N., et al., TGF‐β and basement membrane matrigel stimulate the chondrogenic phenotype in osteoblastic cells derived from fetal rat calvaria. Journal of Bone and Mineral Research, 1996. 11(3): p. 384-391.
91. Xu, C., et al., Feeder-free growth of undifferentiated human embryonic stem cells. Nature biotechnology, 2001. 19(10): p. 971-974.
92. Lu, H., et al., Cultured cell-derived extracellular matrix scaffolds for tissue engineering. Biomaterials, 2011. 32(36): p. 9658-9666.
93. Badylak, S.F., D.O. Freytes, and T.W. Gilbert, Extracellular matrix as a biological scaffold material: Structure and function. Acta biomaterialia, 2009. 5(1): p. 1-13.
94. Acosta, S., et al., Elastin‐Like Recombinamers: Deconstructing and Recapitulating the Functionality of Extracellular Matrix Proteins Using Recombinant Protein Polymers. Advanced Functional Materials, 2020. 30(44): p. 1909050.
95. Jia, J., et al., Evolutionarily conserved sequence motif analysis guides development of chemically defined hydrogels for therapeutic vascularization. Science advances, 2020. 6(28): p. eaaz5894.
96. Park, W.U., et al., A novel vitronectin peptide facilitates differentiation of oligodendrocytes from human pluripotent stem cells (Synthetic ecm for oligodendrocyte differentiation). Biology, 2021. 10(12): p. 1254.
97. Sung, T.-C., et al., Cell-binding peptides on the material surface guide stem cell fate of adhesion, proliferation and differentiation. Journal of Materials Chemistry B, 2023.
98. Sung, T.-C., et al., Poly (vinyl alcohol-co-itaconic acid) hydrogels grafted with several designed peptides for human pluripotent stem cell culture and differentiation into cardiomyocytes. Journal of Materials Chemistry B, 2021. 9(37): p. 7662-7673.
99. Santiago, L.Y., et al., Peptide-surface modification of poly (caprolactone) with laminin-derived sequences for adipose-derived stem cell applications. Biomaterials, 2006. 27(15): p. 2962-2969.
100. 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.
101. Sun, W., et al., Viability and neuronal differentiation of neural stem cells encapsulated in silk fibroin hydrogel functionalized with an IKVAV peptide. Journal of tissue engineering and regenerative medicine, 2017. 11(5): p. 1532-1541.
102. Saleh, N.T., et al., Immobilized laminin-derived peptide can enhance expression of stemness markers in mesenchymal stem cells. Biotechnology and Bioprocess Engineering, 2019. 24: p. 876-884.
103. Li, Y., et al., Aspirin enhances the osteogenic and anti-inflammatory effects of human mesenchymal stem cells on osteogenic BFP-1 peptide-decorated substrates. Journal of Materials Chemistry B, 2017. 5(34): p. 7153-7163.
104. Deng, Y., Y. Yang, and S. Wei, Peptide-decorated nanofibrous niche augments in vitro directed osteogenic conversion of human pluripotent stem cells. Biomacromolecules, 2017. 18(2): p. 587-598.
105. Hayoun‐Neeman, D., et al., Exploring peptide‐functionalized alginate scaffolds for engineering cardiac tissue from human embryonic stem cell‐derived cardiomyocytes in serum‐free medium. Polymers for Advanced Technologies, 2019. 30(10): p. 2493-2505.
106. Yang, Y., Z. Luo, and Y. Zhao, Osteostimulation scaffolds of stem cells: BMP‐7‐derived peptide‐decorated alginate porous scaffolds promote the aggregation and osteo‐differentiation of human mesenchymal stem cells. Biopolymers, 2018. 109(7): p. e23223.
107. Simmons, C.A., et al., Dual growth factor delivery and controlled scaffold degradation enhance in vivo bone formation by transplanted bone marrow stromal cells. Bone, 2004. 35(2): p. 562-569.
108. Jia, J., et al., Development of peptide-functionalized synthetic hydrogel microarrays for stem cell and tissue engineering applications. Acta biomaterialia, 2016. 45: p. 110-120.
109. 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(1): p. 1-16.
110. Jia, J., et al., Engineering alginate as bioink for bioprinting. Acta biomaterialia, 2014. 10(10): p. 4323-4331.
111. Zhou, P., et al., Molecular basis for RGD-containing peptides supporting adhesion and self-renewal of human pluripotent stem cells on synthetic surface. Colloids and Surfaces B: Biointerfaces, 2018. 171: p. 451-460.
112. Pierschbacher, M.D. and E. Ruoslahti, Cell attachment activity of fibronectin can be duplicated by small synthetic fragments of the molecule. Nature, 1984. 309(5963): p. 30-33.
113. Cooke, M., et al., Neural differentiation regulated by biomimetic surfaces presenting motifs of extracellular matrix proteins. Journal of Biomedical Materials Research Part A: An Official Journal of The Society for Biomaterials, The Japanese Society for Biomaterials, and The Australian Society for Biomaterials and the Korean Society for Biomaterials, 2010. 93(3): p. 824-832.
114. Nomizu, M., et al., Cell binding sequences in mouse laminin α1 chain. Journal of Biological Chemistry, 1998. 273(49): p. 32491-32499.
115. Higuchi, A., et al., Polymeric materials for ex vivo expansion of hematopoietic progenitor and stem cells. Journal of Macromolecular Science®, Part C: Polymer Reviews, 2009. 49(3): p. 181-200.
116. Oldberg, A., et al., Identification of a bone sialoprotein receptor in osteosarcoma cells. Journal of Biological Chemistry, 1988. 263(36): p. 19433-19436.
117. Salasznyk, R.M., et al., Adhesion to vitronectin and collagen I promotes osteogenic differentiation of human mesenchymal stem cells. Journal of Biomedicine and Biotechnology, 2004. 2004(1): p. 24-34.
118. Suzuki, S., et al., Complete amino acid sequence of human vitronectin deduced from cDNA. Similarity of cell attachment sites in vitronectin and fibronectin. The EMBO journal, 1985. 4(10): p. 2519-2524.
119. Higuchi, A., et al., Long-term xeno-free culture of human pluripotent stem cells on hydrogels with optimal elasticity. Scientific reports, 2015. 5(1): p. 1-16.
120. 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.
121. Sung, T.-C., et al., Human pluripotent stem cell culture on polyvinyl alcohol-co-itaconic acid hydrogels with varying stiffness under xeno-free conditions. JoVE (Journal of Visualized Experiments), 2018(132): p. e57314.
122. Varun, D., et al., A robust vitronectin-derived peptide for the scalable long-term expansion and neuronal differentiation of human pluripotent stem cell (hPSC)-derived neural progenitor cells (hNPCs). Acta biomaterialia, 2017. 48: p. 120-130.
指導教授 樋口亞紺(Akon Higuchi) 審核日期 2023-8-15
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