博碩士論文 101324010 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:20 、訪客IP:3.139.88.165
姓名 吳孟學(Meng-hsueh Wu)  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 人類脂肪幹細胞培養在具有細胞外基質接枝的水凝膠上之多能性與分化能力研究
(Pluripotency and Differentiation of Human Adipose-derived Stem Cells Cultured on ECM-grafted Hydrogels Having Different Elasticity)
相關論文
★ 連續式培養系統於接枝具奈米鏈段之熱敏感生物材料控制人類胚胎與誘導多功能幹細胞增生★ 重組及培養人類誘導型多能性幹細胞在奈米片段接枝表面
★ 羊水間葉幹細胞培養於細胞外間質寡?嫁接具有硬度/彈性表面的材料,其分化能力及多能性之研究★ 利用具有特定奈米片段及彈性的生醫材料去除癌症幹細胞
★ 從人類初始結腸癌組織分離結腸癌細胞和癌症幹細胞為建立病患專一癌細胞株★ 用於人類胚胎幹細胞生長之生醫材料其奈米片段分子之設計
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) 人類脂肪衍生幹細胞可以經由細胞培養皿來純化,並且顯示具有分化多支鏈的能力。在過去幾十年裡,某些細胞外基質常被塗佈於二維或是三維的幹細胞培養材料上,例如: 膠原蛋白、層黏蛋白質、纖維連接蛋白、玻璃連黏蛋白質以及幹細胞的分化及生長都是常被研究的。然而這些蛋白大多數是由動物衍生出來的所以如果用這些單白質來培養幹細胞,接下來如果要應用於臨床試驗會有異種污染的問題,加上這些蛋白質價格並不便宜。為了實現我們的最終負擔得起的,個性化的再生醫學的目的,臨床上安全和經濟的方式增殖的幹細胞是絕對必要的,同時保持其多能性,以及引導幹細胞譜系規格,而無需使用化學誘導。這裡我研究多能性和人類脂肪衍生幹細胞的分化能力培養於聚(乙烯醇共衣康酸),PVA-IA,水凝膠接枝某些細胞外基質(膠原蛋白第一型,纖連蛋白和寡玻連蛋白)。PVA-IA水凝膠的軟硬度可以透過6小時到48小時的交聯時間加以控制,藉由戊二醛溶液,生成的彈性從2.2千帕至3.7千帕的儲能模量,從E計算對應軟硬度為6.6千帕至11.1千帕的彈性模量= 3G′[生物材料29(2008)2757]。
我們發現人類脂肪衍生幹細胞培養於PVA-IA接枝纖連蛋白及寡玻連蛋白於相對較軟的基材可以維持幹細胞的多能性但是於細胞培養聚苯乙烯的培養皿上卻沒有這個表現,從螢光染色的結果可以發現於我們改質過後的培養皿上有很好的表現但是於細胞培養聚苯乙烯的培養皿卻沒有什麼表現,在成骨分化的分析上
細胞培養聚苯乙烯的培養皿比分化表現優於我們改質過後的培養皿。
摘要(英) Stem cells purified from human adipose tissue (hADSCs) via serial culture of stromal vascular fraction (SVF) on tissue culture plates show multilineage differentiation ability in vitro. In the past decades, extracellular matrices (ECMs) such as collagen, fibronectin, vitronectin, and laminin were used as coating or self-standing materials on 2D culture and 3D culture of stem cells, and the differentiation and proliferation ability of stem cells (e.g., ADSCs or bone marrow-derived mesenchymal stem cells) were investigated. However, the origin of these ECMs are mainly animal-derived or human recombinant, and, therefore, it is not suitable to use them for clinical applications due to xeno-origin contamination and high cost of ECMs. On the other hand, it has been shown that physical environmental factors, such as matrix elasticity, and small functional groups including oligopeptides can have significant effect in determining stem cell fate. To achieve our ultimate goal of affordable, personalized regenerative medicine, clinically safe and economical ways to proliferate stem cells are absolutely essential while maintaining their pluripotency as well as directing stem cell lineage specification without the use of induction chemicals.
Here I investigated pluripotency and differentiation ability of hADSCs cultured on poly(vinylalcohol-co-itaconic acid), PVA-IA, hydrogels grafted with several ECMs (collagen type I, Fibronectin, and oligo-vitronectin [VN]). The stiffness of PVA-IA hydrogels can be controlled by the crosslinking time from 6 hr to 48 hr in glutaraldehyde solution, which generates the elasticity from a 2.2 kPa to 3.7 kPa storage modulus corresponding 6.6 kPa to 11.1 kPa elastic modulus from calculation of E=3G’ [Biomaterials 29 (2008) 2757].
Primary hADSCs were found to maintain their pluripotency on relatively soft hydrogels grafted with Fibronectin and oligopeptide derived from Vitronectin (KGGPQVTRGDVFTMP) from pluripotent gene expression (Nanog, Sox2, Oct4, and klf4) but not on TCPS. From the immunostaining data showed that hADSCs on ECM-grafted PVA-IA films have higher expression of pluripotent proteins than those on TCPS in expansion medium. In the osteogenic differentiation analysis, hADSCs on TCPS have higher differentiation ratio than those on ECM-grafted PVA-IA films after 21 days culture in ostogenic medium.
關鍵字(中) ★ 人類脂肪衍生幹細胞
★ 彈性
★ 間葉幹細胞
關鍵字(英) ★ human adipose derived stem cell
★ elasticity
論文目次 Index of contents
Chapter 1. Introduction 1
1.1 Stem cells 1
1.2 Embryonic stem cells (ESCs) 1
1.2.1 Induced pluripotent stem cells (iPSCs) 2
1.2.2 Hematopoietic stem cells (HSCs) 3
1.2.3 Mesenchymal stem cells (MSCs) 3
1.3 Adipose-derived stem cells 4
1.4 Differentiation capacity of adipose-derived stem cells 6
Environmental factors and bioprocessing parameters that impact on hMSCs fate decision 6
1.4.2 Lineage-specific differentiation potential 10
1.4.3 Immunophenotype 13
1.4.4 Isolation of adipose-derived stem cells 14
1.4.4 Cell isolation 15
1.4.5 Membrane purification method 16
1.4.6 Fluorescence-activated cell sorting (FACS) 17
1.4.7 Magnetic-activated cell sorting (MACS) 20
1.5 ECM Immobilization on 2D dishs 22
1.6 Extracellular matrix (ECM) and nano-segment 24
1.6.1 Type and classification of artificial ECMs 25
1.7 Marker of pluripotent gene 33
1.7.1 Nanog 34
1.7.2 Octamer-binding transcription factor 4 (Oct4) 34
1.7.3 SRY (sex determining region Y)-box 2 (Sox-2) 35
1.7.4 Kruppel-like factor 4 (Klf4) 35
1.8 Marker of differentiation lineages of MSCs 36
1.8.1 Runt-related transcription factor 2 (Runx2) 36
1.8.2 SRY (sex determining region Y)-box 9 (Sox9) 37
1.8.3 Nestin 37
1.8.4 α-actin 38
Chapter 2. Materials and Methods 39
2.1 Materials 39
2.1.1 PVA-IA film 39
2.1.2 Culture medium 39
2.1.3 Supplement in culture medium 39
2.1.4 Digestion solution 40
2.1.5 ACK lysis buffer 40
2.1.6 Phosphate buffer saline solution (PBS) 40
2.1.7 ECM protein 40
2.1.8 RNA extraction 40
2.1.9 Reverse transcriptase (RT) 41
2.1.10 Real-time (qPCR) 41
2.1.11 Probe 41
2.2 Experimental method 43
2.2.1 PVA-IA film preparation 43
2.2.2 Preparation of PVA-IA coating dish grafted with ECM 44
2.2.3 Elasticity measurement of PVA-IA 45
2.2.4 XPS analysis of dish surface 45
2.2.5 10x Phosphate buffer saline (PBS) preparation 45
2.2.6 Culture medium preparation 46
2.2.7 Isolation and culture of adipose tissue-derived stromal cells 46
2.2.8 Culture and passage of ADSCs 49
2.2.9 Cell density measurement 50
2.2.10 Differentiation of adipose tissue-derived stem cells 51
2.2.11 Immunostaining 51
2.2.12 Isolation of total RNA 52
2.2.13 Reverse transcription of mRNA into cDNA 53
2.2.14 Quantitative real time polymerase chain reaction 54
Chapter 3. Results and Discussion 57
3.1 The elasticity of the PVA-IA gel 57
3.2 The XPS measurements of the PVA-IA films 60
3.3 Culture of primary hADSCs on PVA-IA hydrogel grafting collagen having different stiffness 73
3.4 ECM-grafted PVA-IA films having different elasticity guide primary hADSCs into specific lineages of early differentiation 87
3.5 Primary hADSCs culture on crosslinked PVA-IA films (6hr) grafting different ECMs 91
3.6 Different ECM-grafted PVA-IA films guide primary hADSCs into specific lineages of early differentiation 103
Chapter 4 Conclusion 108
Supplement 110
Reference 122










]

Index of Figure
Figure 1-1. Differentiation potential of stem cells. (http://stemcells.nih.gov/info/scireport/pages/chapter1.aspx) 2
Figure 1-2. MSCs have an ability to differentiate into several kinds of cell types. (http://www.genetex.com/WebPage/Product/PromotionDetail.aspx?PromotionID=122) 4
Figure 1-3. Morphology of adipose-derived stem cells. 6
Figure 1-4. Environmental factors and bioprocessing parameters that impact on hPSCs fate decisions (quiescence, self-renewal, differentiation and apoptosis). The main environmental cues and examples of bioprocessing parameters controlling the fate of stem cells are depicted [32]. 7
Figure 1-5. In vitro development of osteoblast phenotype. (http://vschool.scu.edu.tw/biology/content/cytology.htm) 12
Figure 1-6. Cell source of regenerative medicine. 15
Figure 1-7. Preparation procedure of adipose tissue-derived stem cells [26]. 16
Figure 1-8. Diagram of FACS instrument. Cells have been fluorescently tagged with specific antibodies, and the cells that do not express the epitope go to the waste. (http://www.bio.davidson.edu/courses/genomics/method/FACS.html) 20
Figure 1-9. MACS purification of targeted cells after magnetic antibodies labeled. 22
Figure 2-1. The process of making PVA-IA coating dish grafted with ECM. 44
Figure 2-2. The counting grid pattern. (http://www.nexcelom.com/Products/Disposable-Hemacytometer.html) 51
Figure 3-1. The storage modulus of crosslinked PVA-IA films at different angular frequency, which was measured by Rheometer, Physica MCR 101. 58
Figure 3-2. The storage modulus of crosslinked PVA-IA films at1Hz angular frequency, which was measured by Rheometer, Physica MCR 101. 59
Figure 3-3. The storage modulus of crosslinked PVA-IA films at1Hz angular frequency, which was measured by Rheometer, Physica MCR 101. 59
Figure 3-4. The survey spectra of PVA-IA films by X-ray photoelectron spectroscopy. 62
Figure 3-5. The survey spectra of ECM-grafted PVA-IA films by X-ray photoelectron spectroscopy. 63
Figure 3-6. The elemental analysis of PVA-IA films and TCPS by X-ray photoelectron spectroscopy. 64
Figure 3-7. The elemental analysis of PVA-IA films by X-ray photoelectron spectroscopy. 65
Figure 3-8. The elemental analysis of PVA-IA films by X-ray photoelectron spectroscopy. 66
Figure 3-9. The elemental analysis of PVA-IA films by X-ray photoelectron spectroscopy. 67
Figure 3-10. The elemental analysis of ECM-grafted PVA-IA films by X-ray photoelectron spectroscopy. 68
Figure 3-11. The elemental analysis of ECM-grafted PVA-IA films by X-ray photoelectron spectroscopy. 69
Figure 3-12. The elemental analysis of ECM-grafted PVA-IA films by X-ray photoelectron spectroscopy. 70
Figure 3-14. N/C ratio of PVA-IA films, ECM-grafted PVA-IA films and TCPS. 72
Figure 3-15. The microenvironment of primary hADSCs cultured on ECM-grafted PVA-IA films. 75
Figure 3-16. Primary hADSCs culture on ECM-grafted PVA-IA films having different elasticity (e.g., 2.2kPa-3.2kPa). 75
Figure 3-17. The cell morphology of primary hADSCs cultured on PVA-IA grafting dishes having different elasticity (e.g., 2.2kPa ~3.2kPa) and TCPS in expansion medium at Passage 0. The bar indicates 100μm. 76
Figure 3-18. The cell morphology of primary hADSCs cultured on PVA-IA grafting dishes having different elasticity (e.g., 2.2kPa ~3.2kPa) and TCPS in expansion medium at Passage 4. The bar indicates 100μm. 78
Figure 3-19. Doubling time of primary hADSCs cultured on PVA-IA grafting dishes having different elasticity (e.g., 2.2kPa ~3.2kPa) and TCPS in expansion medium at Passage 1. 78
Figure 3-20. Doubling time of primary hADSCs cultured on PVA-IA grafting dishes having different elasticity (e.g., 2.2kPa ~3.2kPa) and TCPS in expansion medium at Passage 4. 79
Figure 3-21. Doubling time of primary hADSCs cultured on PVA-IA grafting dishes having different elasticity (e.g., 2.2kPa ~3.2kPa) and TCPS in expansion medium at Passage 1 and Passage 4. 80
Figure 3-22. The expression of pluripotent gene (Nanog) of primary hADSCs cultured on ECM-grafted PVA-IA films having different elasticity (e.g., 2.2kPa~3.2kPa) in expansion medium at Passage 1. Normalize to Passage 1 TCPS. 81
Figure 3-23. The expression of pluripotent gene (Sox2) of primary hADSCs cultured on ECM-grafted PVA-IA films having different elasticity (e.g., 2.2kPa~3.2kPa) in expansion medium at Passage 1. Normalize to Passage 1 TCPS. 81
Figure 3-24. The expression of pluripotent gene (Oct4) of primary hADSCs cultured on ECM-grafted PVA-IA films having different elasticity (e.g., 2.2kPa~3.2kPa) in expansion medium at Passage 1. Normalize to Passage 1 TCPS. 82
Figure 3-25. The expression of pluripotent gene (klf4) of primary hADSCs cultured on ECM-grafted PVA-IA films having different elasticity (e.g., 2.2kPa~3.2kPa) in expansion medium at Passage 1. Normalize to Passage 1 TCPS. 82
Figure 3-27. The expression of pluripotent gene (Sox2) of primary hADSCs cultured on ECM-grafted PVA-IA films having different elasticity (e.g., 2.2kPa~3.2kPa) in expansion medium at Passage 4. Normalize to Passage 1 TCPS. 83
Figure 3-28. The expression of pluripotent gene (Oct4) of primary hADSCs cultured on ECM-grafted PVA-IA films having different elasticity (e.g., 2.2kPa~3.2kPa) in expansion medium at Passage 4. Normalize to Passage 1 TCPS. 84
Figure 3-29. The expression of pluripotent gene (klf4) of primary hADSCs cultured on ECM-grafted PVA-IA films having different elasticity (e.g., 2.2kPa~3.2kPa) in expansion medium at Passage 4. Normalize to Passage 1 TCPS. 84
Figure 3-30. The expression of pluripotent gene (Nanog) of primary hADSCs cultured on ECM-grafted PVA-IA films having different elasticity (e.g., 2.2kPa~3.2kPa) in expansion medium at Passage 1 and Passage 4. 85
Figure 3-33. The expression of pluripotent gene (klf4) of primary hADSCs cultured on ECM-grafted PVA-IA films having different elasticity (e.g., 2.2kPa~3.2kPa) in expansion medium at Passage 1 and Passage 4. 86
Figure 3-34. The expression of nerve cell gene (Nestin) of primary hADSCs cultured on ECM-grafted PVA-IA films having different elasticity (e.g., 2.2kPa~3.2kPa) in expansion medium at Passage 4. 88
Figure 3-35. The expression of smooth muscle cell gene (α-actin) of primary hADSCs cultured on ECM-grafted PVA-IA films having different elasticity (e.g., 2.2kPa~3.2kPa) in expansion medium at Passage 4. 88
Figure 3-37. The expression of osteogenic gene (Runx2) of primary hADSCs cultured on ECM-grafted PVA-IA films having different elasticity (e.g., 2.2kPa~3.2kPa) in expansion medium at Passage 4. 89
Figure 3-38. The expression of early endoderm differentiation (Sox17) of primary hADSCs cultured on ECM-grafted PVA-IA films having different elasticity (e.g., 2.2kPa~3.2kPa) in expansion medium at Passage 4. 90
Figure 3-39. The relationship between early differentiation genes of three germ layers and different elasticity of ECM-grafted PVA-IA films (e.g., 2.2kPa~3.2kPa) in expansion medium at Passage 4. 90
Figure 3-40. Primary hADSCs culture on ECM-grafted PVA-IA-6hr (e.g., 2.2kPa) films. 92
Figure 3-41. The cell morphology of primary hADSCs culture on PVA-IA-6hr grafting dish and TCPS in expansion medium at Passage 0. The bar indicates 100 m. 93
Figure 3-42. Doubling time of primary hADSCs culture on PVA-IA grafting dish and TCPS in expansion medium at Passage 1. 93
Figure 3-43. The cell morphology of primary hADSCs culture on PVA-IA-6hr grafting dish and TCPS in expansion medium at Passage 4. The bar indicates 100 m. 94
Figure 3-44. Doubling time of primary hADSCs culture on PVA-IA grafting dish and TCPS in expansion medium at Passage 4. 95
Figure 3-45. Doubling time of primary hADSCs culture on PVA-IA grafting dishes and TCPS in expansion medium at Passage 1 and Passage 4. 96
Figure 3-46. The expression of pluripotent gene (Nanog) of primary hADSCs cultured on ECM-grafted PVA-IA films and TCPS in expansion medium at Passage 1. Normalize to Passage 1 TCPS. 96
Figure 3-47. The expression of pluripotent gene (Sox2) of primary hADSCs cultured on ECM-grafted PVA-IA films and TCPS in expansion medium at Passage 1. Normalize to Passage 1 TCPS. 97
Figure 3-48. The expression of pluripotent gene (Oct4) of primary hADSCs cultured on ECM-grafted PVA-IA films and TCPS in expansion medium at Passage 1. Normalize to Passage 1 TCPS. 97
Figure 3-49. The expression of pluripotent gene (klf4) of primary hADSCs cultured on ECM-grafted PVA-IA films and TCPS in expansion medium at Passage 1. Normalize to Passage 1 TCPS. 98
Figure 3-50. The expression of pluripotent gene (Nanog) of primary hADSCs cultured on ECM-grafted PVA-IA films and TCPS in expansion medium at Passage 4. Normalize to Passage 1 TCPS. 98
Figure 3-51. The expression of pluripotent gene (Sox2) of primary hADSCs cultured on ECM-grafted PVA-IA films and TCPS in expansion medium at Passage 4. Normalize to Passage 1 TCPS. 99
Figure 3-52. The expression of pluripotent gene (Oct4) of primary hADSCs cultured on ECM-grafted PVA-IA films and TCPS in expansion medium at Passage 4. Normalize to Passage 1 TCPS. 99
Figure 3-53. The expression of pluripotent gene (klf4) of primary hADSCs cultured on ECM-grafted PVA-IA films and TCPS in expansion medium at Passage 4. Normalize to Passage 1 TCPS. 100
Figure 3-54. The expression of pluripotent gene (Nanog) of primary hADSCs cultured on ECM-grafted PVA-IA films and TCPS in expansion medium at Passage 1 and Passage 4. 100
Figure 3-55. The expression of pluripotent gene (Sox2) of primary hADSCs cultured on ECM-grafted PVA-IA films and TCPS in expansion medium at Passage 1 and Passage 4. 101
Figure 3-56. The expression of pluripotent gene (Oct4) of primary hADSCs cultured on ECM-grafted PVA-IA films and TCPS in expansion medium at Passage 1 and Passage 4. 101
Figure 3-57. The expression of pluripotent gene (klf4) of primary hADSCs cultured on ECM-grafted PVA-IA films and TCPS in expansion medium at Passage 1 and Passage 4. 102
Figure 3-58. Expression of pluripotent markers (Nanog, Sox2) in hADSCs at PVA-IA culture system in expansion medium at Passage 4. The bar indicates 100 m. 102
Figure 3-59. Expression of pluripotent markers (Oct4, SSEA4) of hADSCs at PVA-IA culture system in expansion medium at Passage 4. The bar indicates 100 m. 103
Figure 3-60. The expression of nerve cell gene (Nestin) of primary hADSCs cultured in expansion medium on ECM-grafted PVA-IA films and hES cells culture on matrigel at Passage 4. 104
Figure 3-61. The expression of smooth muscle cell gene (α-actin) of primary hADSCs cultured in expansion medium on ECM-grafted PVA-IA films and hES cells culture on matrigel at Passage 4. 105
Figure 3-62. The expression of early endoderm differentiation (Sox17) of primary hADSCs cultured in expansion medium on ECM-grafted PVA-IA film and hES cells culture on matrigel at Passage 4. 105
Figure 3-63. The relationship between early differentiation genes of three germ layers and different ECM-grafted PVA-IA films cultured in expansion medium at Passage 4. 106
Figure 3-64. The expression of osteogenic differentiation was analyzed by Alizarin Red S staining after the cells had been cultured for 21 days in osteogenic differentiation medium. The bar indicates 100 m. 106
Figure 3-65. The expression of osteogenic differentiation was analyzed by Alizarin Red S staining after the cells had been cultured for 21 days in osteogenic differentiation medium. The bar indicates 100 m. 107







Index of Table
Table 1-1. Characterization of freshly isolated human stromal vascular fractions from adipose tissue and mesenchymal stem cells 14
Table 1-2. Characteristics of common used natural polymers (ECMs) for tissue engineering (modified from Ref. [114]). 26
Table 1-3. Example of characteristics of common used synthetic polymers for tissue engineering (modified from Ref. [114]). 27
Table 1-4 Differentiation markers of MSCs into specific lineages [2]. 36
Table 2-1. Probes of pluripotent gene and different lineage gene. 41
Table 2-2. Primary antibody of immunostaining. 42
Table 2-3. Secondary antibody of immunostaining. 42
Table 2-4 Adipose tissue sample used in this study. 48
Table 2 5 Composition of RNA / primer solution in PCR reaction. 53
Table 2 6 Composition of RT reaction solution. 53
Table 2 7 Composition of qPCR reaction solution. 54
Table 2 8 qPCR step condition. 55
Table 3-1. The result of elasticity of PVA-IA film measured by Rheometer, Physica MCR 101. 60
Table 3-2. O/C ratio of PVA-IA films, ECM-grafted PVA-IA films and TCPS. 71
Table 3-3. N/C ratio of PVA-IA films, ECM-grafted PVA-IA films and TCPS. 72
Table 3-4. Doubling time of primary hADSCs cultured on PVA-IA grafting dishes having different elasticity (e.g., 2.2kPa ~3.2kPa) and TCPS in expansion medium at Passage 1. 79
Table 3-5. Doubling time of primary hADSCs culture on PVA-IA grafting dish having different elasticity (e.g., 2.2kPa ~3.2kPa) and TCPS in expansion medium at Passage 4. 80
Table 3-6. Doubling time of primary hADSCs culture on PVA-IA grafting dish and TCPS in expansion medium at Passage 1. 94
Table 3-7. Doubling time of primary hADSCs culture on PVA-IA grafting dish and TCPS in expansion medium at Passage 4. 95
Table supplement 3-1.The expression of pluripotent gene (Nanog) of primary hADSCs cultured on ECM-grafted PVA-IA films having different elasticity (e.g., 2.2kPa~3.2kPa) in expansion medium at Passage 1. Normalize to Passage 1 TCPS. 110
Table supplement 3-2.The expression of pluripotent gene (Sox2) of primary hADSCs cultured on ECM-grafted PVA-IA films having different elasticity (e.g., 2.2kPa~3.2kPa) in expansion medium at Passage 1. Normalize to Passage 1 TCPS. 110
Table supplement 3-3.The expression of pluripotent gene (Oct4) of primary hADSCs cultured on ECM-grafted PVA-IA films having different elasticity (e.g., 2.2kPa~3.2kPa) in expansion medium at Passage 1. Normalize to Passage 1 TCPS. 110
Table supplement 3-4.The expression of pluripotent gene (klf4) of primary hADSCs cultured on ECM-grafted PVA-IA films having different elasticity (e.g., 2.2kPa~3.2kPa) in expansion medium at Passage 1. Normalize to Passage 1 TCPS. 111
Table supplement 3-5.The expression of pluripotent gene (Nanog) of primary hADSCs cultured on ECM-grafted PVA-IA films having different elasticity (e.g., 2.2kPa~3.2kPa) in expansion medium at Passage 4. Normalize to Passage 1 TCPS. 111
Table supplement 3-6.The expression of pluripotent gene (Sox2) of primary hADSCs cultured on ECM-grafted PVA-IA films having different elasticity (e.g., 2.2kPa~3.2kPa) in expansion medium at Passage 4. Normalize to Passage 1 TCPS. 111
Table supplement 3-7.The expression of pluripotent gene (Oct4) of primary hADSCs cultured on ECM-grafted PVA-IA films having different elasticity (e.g., 2.2kPa~3.2kPa) in expansion medium at Passage 4. Normalize to Passage 1 TCPS. 111
Table supplement 3-8.The expression of pluripotent gene (klf4) of primary hADSCs cultured on ECM-grafted PVA-IA films having different elasticity (e.g., 2.2kPa~3.2kPa) in expansion medium at Passage 4. Normalize to Passage 1 TCPS. 112
Table supplement 3-9.The expression of nerve cell gene (Nestin) of primary hADSCs cultured on ECM-grafted PVA-IA films having different elasticity (e.g., 2.2kPa~3.2kPa) in expansion medium at Passage 4. 112
Table supplement 3-10.The expression of smooth muscle cell gene (α-actin) of primary hADSCs cultured on ECM-grafted PVA-IA films having different elasticity (e.g., 2.2kPa~3.2kPa) in expansion medium at Passage 4. 112
Table supplement 3-11.The expression of chondrocyte gene (Sox9) of primary hADSCs cultured on ECM-grafted PVA-IA films having different elasticity (e.g., 2.2kPa~3.2kPa) in expansion medium at Passage 4. 113
Table supplement 3-12.The expression of osteogenic gene (Runx2) of primary hADSCs cultured on ECM-grafted PVA-IA films having different elasticity (e.g., 2.2kPa~3.2kPa) in expansion medium at Passage 4. 113
Table supplement 3-13.The expression of early endoderm differentiation (Sox17) of primary hADSCs cultured on ECM-grafted PVA-IA films having different elasticity (e.g., 2.2kPa~3.2kPa) in expansion medium at Passage 4. 113
Table supplement 3-14.The expression of pluripotent gene (Nanog) of primary hADSCs cultured on ECM-grafted PVA-IA films and TCPS in expansion medium at Passage 1. Normalize to Passage 1 TCPS. 114
Table supplement 3-15.The expression of pluripotent gene (Sox2) of primary hADSCs cultured on ECM-grafted PVA-IA films and TCPS in expansion medium at Passage 1. Normalize to Passage 1 TCPS. 114
Table supplement 3-16. The expression of pluripotent gene (Oct4) of primary hADSCs cultured on ECM-grafted PVA-IA films and TCPS in expansion medium at Passage 1. Normalize to Passage 1 TCPS. 114
Table supplement 3-17. The expression of pluripotent gene (klf4) of primary hADSCs cultured on ECM-grafted PVA-IA films and TCPS in expansion medium at Passage 1. Normalize to Passage 1 TCPS. 114
Table supplement 3-18.The expression of pluripotent gene (Nanog) of primary hADSCs cultured on ECM-grafted PVA-IA films and TCPS in expansion medium at Passage 4. Normalize to Passage 1 TCPS. 115
Table supplement 3-19.The expression of pluripotent gene (Sox2) of primary hADSCs cultured on ECM-grafted PVA-IA films and TCPS in expansion medium at Passage 4. Normalize to Passage 1 TCPS. 115
Table supplement 3-20.The expression of pluripotent gene (Oct4) of primary hADSCs cultured on ECM-grafted PVA-IA films and TCPS in expansion medium at Passage 4. Normalize to Passage 1 TCPS. 115
Table supplement 3-21.The expression of pluripotent gene (klf4) of primary hADSCs cultured on ECM-grafted PVA-IA films and TCPS in expansion medium at Passage 4. Normalize to Passage 1 TCPS. 115
Table supplement 3-22.The expression of nerve cell gene (Nestin) of primary hADSCs cultured in expansion medium on ECM-grafted PVA-IA films and hES cells culture on matrigel at Passage 4. 116
Table supplement 3-23.The expression of smooth muscle cell gene (α-actin) of primary hADSCs cultured in expansion medium on ECM-grafted PVA-IA films and hES cells culture on matrigel at Passage 4. 116
Table supplement 3-24.The expression of early endoderm differentiation (Sox17) of primary hADSCs cultured in expansion medium on ECM-grafted PVA-IA film and hES cells culture on matrigel at Passage 4. 116
Table supplement 3-25.The expression of osteogenic differentiation was analyzed by Alizarin Red S staining after the cells had been cultured for 21 days in osteogenic differentiation medium. 116
Table supplement 3-26.The expression of pluripotent gene (Nanog) of primary hADSCs cultured on ECM-grafted PVA-IA films having different elasticity (e.g., 2.2kPa~3.2kPa) in expansion medium at Passage 1 and Passage 4. 117
Table supplement 3-27.The expression of pluripotent gene (Sox2) of primary hADSCs cultured on ECM-grafted PVA-IA films having different elasticity (e.g., 2.2kPa~3.2kPa) in expansion medium at Passage 1 and Passage 4. 117
Table supplement 3-28.The expression of pluripotent gene (Oct4) of primary hADSCs cultured on ECM-grafted PVA-IA films having different elasticity (e.g., 2.2kPa~3.2kPa) in expansion medium at Passage 1 and Passage 4. 118
Table supplement 3-29.The expression of pluripotent gene (klf4) of primary hADSCs cultured on ECM-grafted PVA-IA films having different elasticity (e.g., 2.2kPa~3.2kPa) in expansion medium at Passage 1 and Passage 4. 118
Table supplement 3-30.The expression of pluripotent gene (Nanog) of primary hADSCs cultured on ECM-grafted PVA-IA films and TCPS in expansion medium at Passage 1 and Passage 4. 118
Table supplement 3-31.The expression of pluripotent gene (Sox2) of primary hADSCs cultured on ECM-grafted PVA-IA films and TCPS in expansion medium at Passage 1 and Passage 4. 119
Table supplement 3-32.The expression of pluripotent gene (Oct4) of primary hADSCs cultured on ECM-grafted PVA-IA films and TCPS in expansion medium at Passage 1 and Passage 4. 119
Table supplement 3-33.The expression of pluripotent gene (klf4) of primary hADSCs cultured on ECM-grafted PVA-IA films and TCPS in expansion medium at Passage 1 and Passage 4. 119
Table supplement 3-34. Doubling time of primary hADSCs culture on PVA-IA grafting dishes and TCPS in expansion medium at Passage 1 and Passage 4. 120
Table supplement 3-35. Doubling time of primary hADSCs culture on PVA-IA grafting dishes and TCPS in expansion medium at Passage 1 and Passage 4. 120
Table supplement 3-36.The expression of early differentiation marker of primary hADSCs cultured in expansion medium on ECM-grafted PVA-IA films and TCPS at Passage 4. 120
Table supplement 3-37.The expression of early differentiation marker of primary hADSCs cultured in expansion medium on ECM-grafted PVA-IA films, TCPS and hES on matrigel at Passage 4. 121


參考文獻 1. Jiang, Y.H., et al., Pluripotency of mesenchymal stem cells derived from adult marrow (vol 418, pg 41, 2002). Nature, 2007. 447(7146): p. 879-880.
2. Higuchi, A., et al., Biomimetic Cell Culture Proteins as Extracellular Matrices for Stem Cell Differentiation. Chemical Reviews, 2012. 112(8): p. 4507-4540.
3. Okita, K., T. Ichisaka, and S. Yamanaka, Generation of germline-competent induced pluripotent stem cells. Nature, 2007. 448(7151): p. 313-7.
4. 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.
5. Yu, J., et al., Induced pluripotent stem cell lines derived from human somatic cells. Science, 2007. 318(5858): p. 1917-20.
6. Lin, S.L., et al., Mir-302 reprograms human skin cancer cells into a pluripotent ES-cell-like state. RNA, 2008. 14(10): p. 2115-24.
7. Zhou, H., et al., Generation of induced pluripotent stem cells using recombinant proteins. Cell Stem Cell, 2009. 4(5): p. 381-4.
8. Favre, G., et al., Differences between graft product and donor side effects following bone marrow or stem cell donation. Bone Marrow Transplant, 2003. 32(9): p. 873-80.
9. Bosi, A. and B. Bartolozzi, Safety of bone marrow stem cell donation: a review. Transplant Proc, 2010. 42(6): p. 2192-4.
10. Gratwohl, A., et al., Predictability of hematopoietic stem cell transplantation rates. Haematologica, 2007. 92(12): p. 1679-86.
11. Hamidieh, A.A., et al., Autologous stem cell transplantation as treatment modality in a patient with relapsed pancreatoblastoma. Pediatr Blood Cancer, 2010. 55(3): p. 573-6.
12. Higuchi, A., et al., Separation of hematopoietic stem cells from human peripheral blood through modified polyurethane foaming membranes. Journal of Biomedical Materials Research Part A, 2008. 85A(4): p. 853-861.
13. Caplan, A.I. and J.E. Dennis, Mesenchymal stem cells as trophic mediators. J Cell Biochem, 2006. 98(5): p. 1076-84.
14. Scadden, D.T., The stem-cell niche as an entity of action. Nature, 2006. 441(7097): p. 1075-1079.
15. Horwitz, E.M., et al., Isolated allogeneic bone marrow-derived mesenchymal cells engraft and stimulate growth in children with osteogenesis imperfecta: Implications for cell therapy of bone. Proc Natl Acad Sci U S A, 2002. 99(13): p. 8932-7.
16. Arinzeh, T.L., et al., Allogeneic mesenchymal stem cells regenerate bone in a critical-sized canine segmental defect. J Bone Joint Surg Am, 2003. 85-A(10): p. 1927-35.
17. Pittenger, M.F., et al., Multilineage potential of adult human mesenchymal stem cells. Science, 1999. 284(5411): p. 143-147.
18. Kern, S., et al., Comparative analysis of mesenchymal stem cells from bone marrow, umbilical cord blood, or adipose tissue. Stem Cells, 2006. 24(5): p. 1294-301.
19. Crisan, M., et al., A perivascular origin for mesenchymal stem cells in multiple human organs. Cell Stem Cell, 2008. 3(3): p. 301-13.
20. Brighton, C.T. and R.M. Hunt, Early histological and ultrastructural changes in medullary fracture callus. J Bone Joint Surg Am, 1991. 73(6): p. 832-47.
21. Netter, F.H., Musculoskeletal system : anatomy, physiology, and metabolic disorders. U.S.A : Indoo. 1987.
22. Zuk, P.A., et al., Multilineage cells from human adipose tissue: implications for cell-based therapies. Tissue Eng, 2001. 7(2): p. 211-28.
23. Mizuno, H., M. Tobita, and A.C. Uysal, Concise review: Adipose-derived stem cells as a novel tool for future regenerative medicine. Stem Cells, 2012. 30(5): p. 804-10.
24. Zuk, P.A., et al., Human adipose tissue is a source of multipotent stem cells. Mol Biol Cell, 2002. 13(12): p. 4279-95.
25. van Dijk, A., et al., Differentiation of human adipose-derived stem cells towards cardiomyocytes is facilitated by laminin. Cell Tissue Res, 2008. 334(3): p. 457-67.
26. Schaffler, A. and C. Buchler, Concise review: adipose tissue-derived stromal cells--basic and clinical implications for novel cell-based therapies. Stem Cells, 2007. 25(4): p. 818-27.
27. Oedayrajsingh-Varma, M.J., et al., Adipose tissue-derived mesenchymal stem cell yield and growth characteristics are affected by the tissue-harvesting procedure. Cytotherapy, 2006. 8(2): p. 166-77.
28. Mitchell, J.B., et al., Immunophenotype of human adipose-derived cells: temporal changes in stromal-associated and stem cell-associated markers. Stem Cells, 2006. 24(2): p. 376-85.
29. Zaragosi, L.E., G. Ailhaud, and C. Dani, Autocrine fibroblast growth factor 2 signaling is critical for self-renewal of human multipotent adipose-derived stem cells. Stem Cells, 2006. 24(11): p. 2412-9.
30. Rubio, D., et al., Spontaneous human adult stem cell transformation. Cancer Res, 2005. 65(8): p. 3035-9.
31. Azarin, S.M. and S.P. Palecek, Development of scalable culture systems for human embryonic stem cells. Biochemical Engineering Journal, 2010. 48(3): p. 378-384.
32. Serra, M., et al., Process engineering of human pluripotent stem cells for clinical application. Trends in Biotechnology, 2012. 30(6): p. 350-359.
33. 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.
34. Kolhar, P., et al., Synthetic surfaces for human embryonic stem cell culture. Journal of Biotechnology, 2010. 146(3): p. 143-146.
35. 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.
36. Ferreira, L.S., et al., Bioactive hydrogel scaffolds for controllable vascular differentiation of human embryonic stem cells. Biomaterials, 2007. 28(17): p. 2706-2717.
37. Maia, J., et al., Controlling the Neuronal Differentiation of Stem Cells by the Intracellular Delivery of Retinoic Acid-Loaded Nanoparticles. Acs Nano, 2011. 5(1): p. 97-106.
38. Ao, A., J.J. Hao, and C.C. Hong, Regenerative Chemical Biology: Current Challenges and Future Potential. Chemistry & Biology, 2011. 18(4): p. 413-424.
39. Burdick, J.A. and F.M. Watt, High-throughput stem-cell niches. Nature Methods, 2011. 8(11): p. 915-916.
40. Bauwens, C.L., et al., Control of human embryonic stem cell colony and aggregate size heterogeneity influences differentiation trajectories. Stem Cells, 2008. 26(9): p. 2300-2310.
41. Engler, A.J., et al., Matrix elasticity directs stem cell lineage specification. Cell, 2006. 126(4): p. 677-689.
42. Veraitch, F.S., et al., The impact of manual processing on the expansion and directed differentiation of embryonic stem cells. Biotechnology and Bioengineering, 2008. 99(5): p. 1216-1229.
43. Ezashi, T., P. Das, and R.M. Roberts, Low O2 tensions and the prevention of differentiation of hES cells. Proceedings of the National Academy of Sciences of the United States of America, 2005. 102(13): p. 4783-4788.
44. Forsyth, N.R., et al., Physiologic oxygen enhances human embryonic stem cell clonal recovery and reduces chromosomal abnormalities. Cloning and Stem Cells, 2006. 8(1): p. 16-23.
45. Liu, Q., et al., A comparative study of proliferation and osteogenic differentiation of adipose-derived stem cells on akermanite and beta-TCP ceramics. Biomaterials, 2008. 29(36): p. 4792-9.
46. Ahn, H.H., et al., In vivo osteogenic differentiation of human adipose-derived stem cells in an injectable in situ-forming gel scaffold. Tissue Eng Part A, 2009. 15(7): p. 1821-32.
47. Flynn, L.E., The use of decellularized adipose tissue to provide an inductive microenvironment for the adipogenic differentiation of human adipose-derived stem cells. Biomaterials, 2010. 31(17): p. 4715-24.
48. Natesan, S., et al., Adipose-Derived Stem Cell Delivery into Collagen Gels Using Chitosan Microspheres. Tissue Engineering Part A, 2010. 16(4): p. 1369-1384.
49. Awad, H.A., et al., Chondrogenic differentiation of adipose-derived adult stem cells in agarose, alginate, and gelatin scaffolds. Biomaterials, 2004. 25(16): p. 3211-3222.
50. Betre, H., et al., Chondrocytic differentiation of human adipose-derived adult stem cells in elastin-like polypeptide. Biomaterials, 2006. 27(1): p. 91-99.
51. Estes, B.T., A.W. Wu, and F. Guilak, Potent induction of chondrocytic differentiation of human adipose-derived adult stem cells by bone morphogenetic protein 6. Arthritis and Rheumatism, 2006. 54(4): p. 1222-1232.
52. Mahmoudifar, N. and P.M. Doran, Chondrogenic differentiation of human adipose-derived stem cells in polyglycolic acid mesh scaffolds under dynamic culture conditions. Biomaterials, 2010. 31(14): p. 3858-3867.
53. van Dijk, A., et al., Differentiation of human adipose-derived stem cells towards cardiomyocytes is facilitated by laminin. Cell and Tissue Research, 2008. 334(3): p. 457-467.
54. Madonna, R. and R. De Caterina, Adipose tissue: a new source for cardiovascular repair. Journal of Cardiovascular Medicine, 2010. 11(2): p. 71-80.
55. Huang, T.T., et al., Neuron-like differentiation of adipose-derived stem cells from infant piglets in vitro. Journal of Spinal Cord Medicine, 2007. 30: p. S35-S40.
56. Kingham, P.J., C. Mantovani, and G. Terenghi, Notch independent signalling mediates Schwann cell-like differentiation of Adipose Derived Stem Cells. Neuroscience Letters, 2009. 467(2): p. 164-168.
57. Zemel, R., et al., Expression of liver-specific markers in naive adipose-derived mesenchymal stem cells. Liver International, 2009. 29(9): p. 1326-1337.
58. Timper, K., et al., Human adipose tissue-derived mesenchymal stem cells differentiate into insulin, somatostatin, and glucagon expressing cells. Biochemical and Biophysical Research Communications, 2006. 341(4): p. 1135-1140.
59. Chandra, V., et al., Generation of Pancreatic Hormone-Expressing Islet-Like Cell Aggregates from Murine Adipose Tissue-Derived Stem Cells. Stem Cells, 2009. 27(8): p. 1941-1953.
60. Xu, M.G., et al., An cell-assembly derived physiological 3D model of the metabolic syndrome, based on adipose-derived stromal cells and a gelatin/alginate/fibrinogen matrix. Biomaterials, 2010. 31(14): p. 3868-3877.
61. von Heimburg, D., et al., Human preadipocytes seeded on freeze-dried collagen scaffolds investigated in vitro and in vivo. Biomaterials, 2001. 22(5): p. 429-438.
62. Halbleib, M., et al., Tissue engineering of white adipose tissue using hyaluronic acid-based scaffolds. I: in vitro differentiation of human adipocyte precursor cells on scaffolds. Biomaterials, 2003. 24(18): p. 3125-3132.
63. Dawson, J.I., et al., Development of specific collagen scaffolds to support the osteogenic and chondrogenic differentiation of human bone marrow stromal cells. Biomaterials, 2008. 29(21): p. 3105-3116.
64. Park, I.S., et al., The correlation between human adipose-derived stem cells differentiation and cell adhesion mechanism. Biomaterials, 2009. 30(36): p. 6835-6843.
65. Nigro, J., et al., The effect of bovine endosteum-derived particles on the proliferation of human mesenchymal stem cells. Biomaterials, 2010. 31(21): p. 5689-5699.
66. Ward, D.F., Jr., et al., Mechanical strain enhances extracellular matrix-induced gene focusing and promotes osteogenic differentiation of human mesenchymal stem cells through an extracellular-related kinase-dependent pathway. Stem Cells and Development, 2007. 16(3): p. 467-479.
67. Rider, D.A., et al., Autocrine fibroblast growth factor 2 increases the multipotentiality of human adipose-derived mesenchymal stem cells. Stem Cells, 2008. 26(6): p. 1598-608.
68. Mochizuki, T., et al., Higher chondrogenic potential of fibrous synovium- and adipose synovium-derived cells compared with subcutaneous fat-derived cells: distinguishing properties of mesenchymal stem cells in humans. Arthritis Rheum, 2006. 54(3): p. 843-53.
69. Stein, G.S., et al., Transcriptional control of osteoblast growth and differentiation. Physiol Rev, 1996. 76(2): p. 593-629.
70. Martin, I., et al., Selective differentiation of mammalian bone marrow stromal cells cultured on three-dimensional polymer foams. J Biomed Mater Res, 2001. 55(2): p. 229-35.
71. Noth, U., et al., Chondrogenic differentiation of human mesenchymal stem cells in collagen type I hydrogels. J Biomed Mater Res A, 2007. 83(3): p. 626-35.
72. Planat-Benard, V., et al., Spontaneous cardiomyocyte differentiation from adipose tissue stroma cells. Circ Res, 2004. 94(2): p. 223-9.
73. Miyahara, Y., et al., Monolayered mesenchymal stem cells repair scarred myocardium after myocardial infarction. Nat Med, 2006. 12(4): p. 459-65.
74. Strem, B.M., et al., Expression of cardiomyocytic markers on adipose tissue-derived cells in a murine model of acute myocardial injury. Cytotherapy, 2005. 7(3): p. 282-91.
75. Rodriguez, A.M., et al., Transplantation of a multipotent cell population from human adipose tissue induces dystrophin expression in the immunocompetent mdx mouse. J Exp Med, 2005. 201(9): p. 1397-405.
76. Zimmerlin, L., et al., Stromal vascular progenitors in adult human adipose tissue. Cytometry A, 2010. 77(1): p. 22-30.
77. Yoshimura, K., et al., Characterization of freshly isolated and cultured cells derived from the fatty and fluid portions of liposuction aspirates. J Cell Physiol, 2006. 208(1): p. 64-76.
78. Civin, C.I., et al., Antigenic analysis of hematopoiesis. III. A hematopoietic progenitor cell surface antigen defined by a monoclonal antibody raised against KG-1a cells. J Immunol, 1984. 133(1): p. 157-65.
79. Asahara, T., et al., Isolation of putative progenitor endothelial cells for angiogenesis. Science, 1997. 275(5302): p. 964-7.
80. Pusztaszeri, M.P., W. Seelentag, and F.T. Bosman, Immunohistochemical expression of endothelial markers CD31, CD34, von Willebrand factor, and Fli-1 in normal human tissues. J Histochem Cytochem, 2006. 54(4): p. 385-95.
81. Lin, G., et al., Defining stem and progenitor cells within adipose tissue. Stem Cells Dev, 2008. 17(6): p. 1053-63.
82. Traktuev, D.O., et al., A population of multipotent CD34-positive adipose stromal cells share pericyte and mesenchymal surface markers, reside in a periendothelial location, and stabilize endothelial networks. Circ Res, 2008. 102(1): p. 77-85.
83. Zannettino, A.C., et al., Multipotential human adipose-derived stromal stem cells exhibit a perivascular phenotype in vitro and in vivo. J Cell Physiol, 2008. 214(2): p. 413-21.
84. Sengenes, C., et al., Preadipocytes in the human subcutaneous adipose tissue display distinct features from the adult mesenchymal and hematopoietic stem cells. J Cell Physiol, 2005. 205(1): p. 114-22.
85. Higuchi, A., et al., Cell separation between mesenchymal progenitor cells through porous polymeric membranes. J Biomed Mater Res B Appl Biomater, 2005. 74(1): p. 511-9.
86. Higuchi, A., et al., Separation of CD34+ cells from human peripheral blood through polyurethane foaming membranes. J Biomed Mater Res A, 2006. 78(3): p. 491-9.
87. Rodbell, M., Metabolism of isolated fat cells. II. The similar effects of phospholipase C (Clostridium perfringens alpha toxin) and of insulin on glucose and amino acid metabolism. J Biol Chem, 1966. 241(1): p. 130-9.
88. Rodbell, M., The metabolism of isolated fat cells. IV. Regulation of release of protein by lipolytic hormones and insulin. J Biol Chem, 1966. 241(17): p. 3909-17.
89. Rodbell, M. and A.B. Jones, Metabolism of isolated fat cells. 3. The similar inhibitory action of phospholipase C (Clostridium perfringens alpha toxin) and of insulin on lipolysis stimulated by lipolytic hormones and theophylline. J Biol Chem, 1966. 241(1): p. 140-2.
90. Kurita, M., et al., Influences of centrifugation on cells and tissues in liposuction aspirates: optimized centrifugation for lipotransfer and cell isolation. Plast Reconstr Surg, 2008. 121(3): p. 1033-41; discussion 1042-3.
91. Chen, D.-C., et al., Purification of human adipose-derived stem cells from fat tissues using PLGA/silk screen hybrid membranes. Biomaterials, 2014. 35(14): p. 4278-4287.
92. Higuchi, A., et al., Peripheral blood cell separation through surface-modified polyurethane membranes. J Biomed Mater Res A, 2004. 68(1): p. 34-42.
93. Higuchi, A., et al., Separation of Hematopoietic Stem and Progenitor Cells from Human Peripheral Blood Through Polyurethane Foaming Membranes Modified with Several Amino Acids. Journal of Applied Polymer Science, 2009. 114(2): p. 671-679.
94. Wu, C.H., et al., The isolation and differentiation of human adipose-derived stem cells using membrane filtration. Biomaterials, 2012. 33(33): p. 8228-39.
95. Fulwyler, M.J., Electronic separation of biological cells by volume. Science, 1965. 150(3698): p. 910-1.
96. Sweet, R.G., High Frequency Recording with Electrostatically Deflected Ink Jets. Review of Scientific Instruments, 1965. 36(2): p. 131-136.
97. Van Dilla, M.A., M.J. Fulwyler, and I.U. Boone, Volume Distribution and Separation of Normal Human Leucocytes. Experimental Biology and Medicine, 1967. 125(2): p. 367-370.
98. Bonner, W.A., et al., Fluorescence Activated Cell Sorting. Review of Scientific Instruments, 1972. 43(3): p. 404-409.
99. Johnson, K.W., M. Dooner, and P.J. Quesenberry, Fluorescence Activated Cell Sorting: A Window on the Stem Cell. Current Pharmaceutical Biotechnology, 2007. 8(3): p. 133-139.
100. Assenmacher, M., et al., FLUORESCENCE-ACTIVATED CYTOMETRY CELL SORTING BASED ON IMMUNOLOGICAL RECOGNITION. Clinical Biochemistry, 1995. 28(1): p. 39-40.
101. Miltenyi, S., et al., High gradient magnetic cell separation with MACS. Cytometry, 1990. 11(2): p. 231-238.
102. Kato, K. and A. Radbruch, Isolation and characterization of CD34+ hematopoietic stem cells from human peripheral blood by high-gradient magnetic cell sorting. Cytometry, 1993. 14(4): p. 384-392.
103. de Wynter, E.A., et al., Comparison of purity and enrichment of CD34+ cells from bone marrow, umbilical cord and peripheral blood (primed for apheresis) using five separation systems. Stem Cells, 1995. 13(5): p. 524-32.
104. McNiece, I., et al., Large-scale isolation of CD34+ cells using the Amgen cell selection device results in high levels of purity and recovery. J Hematother, 1997. 6(1): p. 5-11.
105. Richel, D.J., et al., Highly purified CD34+ cells isolated using magnetically activated cell selection provide rapid engraftment following high-dose chemotherapy in breast cancer patients. Bone Marrow Transplant, 2000. 25(3): p. 243-9.
106. Rosso, F., et al., From cell-ECM interactions to tissue engineering. J Cell Physiol, 2004. 199(2): p. 174-80.
107. Langer, R. and J.P. Vacanti, Tissue engineering. Science, 1993. 260(5110): p. 920-6.
108. Putnam, A.J. and D.J. Mooney, Tissue engineering using synthetic extracellular matrices. Nature Medicine, 1996. 2(7): p. 824-826.
109. Daley, W.P., S.B. Peters, and M. Larsen, Extracellular matrix dynamics in development and regenerative medicine. Journal of Cell Science, 2008. 121(3): p. 255-264.
110. Rozario, T. and D.W. DeSimone, The extracellular matrix in development and morphogenesis: A dynamic view. Developmental Biology, 2010. 341(1): p. 126-140.
111. Chen, L.Y., et al., Effect of the surface density of nanosegments immobilized on culture dishes on ex vivo expansion of hematopoietic stem and progenitor cells from umbilical cord blood. Acta Biomaterialia, 2012. 8(5): p. 1749-1758.
112. Rosso, F., et al., Smart materials as scaffolds for tissue engineering. Journal of Cellular Physiology, 2005. 203(3): p. 465-470.
113. Moroni, L., J.R. De Wijn, and C.A. Van Blitterswijk, Integrating novel technologies to fabricate smart scaffolds. Journal of Biomaterials Science-Polymer Edition, 2008. 19(5): p. 543-572.
114. Mano, J.F., et al., Natural origin biodegradable systems in tissue engineering and regenerative medicine: present status and some moving trends. Journal of the Royal Society Interface, 2007. 4(17): p. 999-1030.
115. Di Lullo, G.A., et al., Mapping the ligand-binding sites and disease-associated mutations on the most abundant protein in the human, type I collagen. Journal of Biological Chemistry, 2002. 277(6): p. 4223-4231.
116. Hosseinkhani, H., et al., Perfusion culture enhances osteogenic differentiation of rat mesenchymal stem cells in collagen sponge reinforced with poly( glycolic acid) fiber. Tissue Engineering, 2005. 11(9-10): p. 1476-1488.
117. Shih, Y.R.V., et al., Growth of mesenchymal stem cells on electrospun type I collagen nanofibers. Stem Cells, 2006. 24(11): p. 2391-2397.
118. Donzelli, E., et al., Mesenchymal stem cells cultured on a collagen scaffold: In vitro osteogenic differentiation. Archives of Oral Biology, 2007. 52(1): p. 64-73.
119. Sefcik, L.S., et al., Collagen nanofibres are a biomimetic substrate for the serum-free osteogenic differentiation of human adipose stem cells. Journal of Tissue Engineering and Regenerative Medicine, 2008. 2(4): p. 210-220.
120. Mueller, S.M. and J. Glowacki, Age-related decline in the osteogenic potential of human bone marrow cells cultured in three-dimensional collagen sponges. Journal of Cellular Biochemistry, 2001. 82(4): p. 583-590.
121. Sado, Y., et al., Organization and expression of basement membrane collagen IV genes and their roles in human disorders. Journal of Biochemistry, 1998. 123(5): p. 767-776.
122. Giunta, C., et al., Homozygous Gly530Ser substitution in COL5A1 causes mild classical Ehlers-Danlos syndrome. American Journal of Medical Genetics, 2002. 109(4): p. 284-290.
123. Fernandes, H., et al., The Role of Collagen Crosslinking in Differentiation of Human Mesenchymal Stem Cells and MC3T3-E1 Cells. Tissue Engineering Part A, 2009. 15(12): p. 3857-3867.
124. Ma, W., et al., CNS stem and progenitor cell differentiation into functional neuronal circuits in three-dimensional collagen gels. Experimental Neurology, 2004. 190(2): p. 276-288.
125. Mao, Y. and J.E. Schwarzbauer, Fibronectin fibrillogenesis, a cell-mediated matrix assembly process. Matrix Biology, 2005. 24(6): p. 389-399.
126. Drake, S.L., et al., Structural Features of Fibronectin Synthetic Peptide Fn-C/H Ii, Responsible for Cell-Adhesion, Neurite Extension, and Heparan-Sulfate Binding. Journal of Biological Chemistry, 1993. 268(21): p. 15859-15867.
127. Manton, K.J., et al., Disruption of heparan and chondroitin sulfate signaling enhances mesenchymal stem cell-derived osteogenic differentiation via bone morphogenetic protein signaling pathways. Stem Cells, 2007. 25(11): p. 2845-2854.
128. McBeath, R., et al., Cell shape, cytoskeletal tension, and RhoA regulate stem cell lineage commitment. Developmental Cell, 2004. 6(4): p. 483-495.
129. Chang, C.F., et al., Fibronectin and pellet suspension culture promote differentiation of human mesenchymal stem cells into insulin producing cells. Journal of Biomedical Materials Research Part A, 2008. 86A(4): p. 1097-1105.
130. Jiang, F.X., et al., Laminin-1 promotes differentiation of fetal mouse pancreatic beta-cells. Diabetes, 1999. 48(4): p. 722-730.
131. Sogo, Y., et al., Fibronectin-calcium phosphate composite layer on hydroxyapatite to enhance adhesion, cell spread and osteogenic differentiation of human mesenchymal stem cells in vitro. Biomedical Materials, 2007. 2(2): p. 116-123.
132. Kikkawa, Y., et al., Integrin binding specificity of laminin-10/11 : laminin-10/11 are recognized by alpha 3 beta 1, alpha 6 beta 1 and alpha 6 beta 4 integrins. Journal of Cell Science, 2000. 113(5): p. 869-876.
133. Ogawa, T., et al., The short arm of laminin gamma 2 chain of laminin-5 (laminin-332) binds syndecan-1 and regulates cellular adhesion and migration by suppressing phosphorylation of integrin beta 4 chain. Molecular Biology of the Cell, 2007. 18(5): p. 1621-1633.
134. Klees, R.F., et al., Laminin-5 induces osteogenic gene expression in human mesenchymal stem cells through an ERK-dependent pathway. Molecular Biology of the Cell, 2005. 16(2): p. 881-890.
135. Suzuki, S., et al., Effects of Extracellular Matrix on Differentiation of Human Bone Marrow-Derived Mesenchymal Stem Cells into Smooth Muscle Cell Lineage: Utility for Cardiovascular Tissue Engineering. Cells Tissues Organs, 2010. 191(4): p. 269-280.
136. Gil, J.E., et al., Vitronectin promotes oligodendrocyte differentiation during neurogenesis of human embryonic stem cells. Febs Letters, 2009. 583(3): p. 561-567.
137. Takebayashi, H., et al., The basic helix-loop-helix factor Olig2 is essential for the development of motoneuron and oligodendrocyte lineages. Current Biology, 2002. 12(13): p. 1157-1163.
138. 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(1): p. 24-34.
139. Kleinman, H.K. and G.R. Martin, Matrigel: basement membrane matrix with biological activity. Semin Cancer Biol, 2005. 15(5): p. 378-86.
140. Donovan, P.J. and J. Gearhart, The end of the beginning for pluripotent stem cells. Nature, 2001. 414(6859): p. 92-97.
141. Rosner, M.H., et al., A Pou-Domain Transcription Factor in Early Stem-Cells and Germ-Cells of the Mammalian Embryo. Nature, 1990. 345(6277): p. 686-692.
142. Carlin, R., et al., Expression of early transcription factors Oct-4, Sox-2 and Nanog by porcine umbilical cord (PUC) matrix cells. Reproductive Biology and Endocrinology, 2006. 4.
143. Chambers, I., et al., Functional expression cloning of Nanog, a pluripotency sustaining factor in embryonic stem cells. Cell, 2003. 113(5): p. 643-55.
144. Zaehres, H., et al., High-Efficiency RNA Interference in Human Embryonic Stem Cells. STEM CELLS, 2005. 23(3): p. 299-305.
145. Takeda, J., S. Seino, and G.I. Bell, Human Oct3 gene family: cDNA sequences, alternative splicing, gene organization, chromosomal location, and expression at low levels in adult tissues. Nucleic Acids Res, 1992. 20(17): p. 4613-20.
146. Rodda, D.J., et al., Transcriptional regulation of nanog by OCT4 and SOX2. J Biol Chem, 2005. 280(26): p. 24731-7.
147. Masui, S., et al., Pluripotency governed by Sox2 via regulation of Oct3/4 expression in mouse embryonic stem cells. Nat Cell Biol, 2007. 9(6): p. 625-35.
148. Rowland, B.D. and D.S. Peeper, KLF4, p21 and context-dependent opposing forces in cancer. Nat Rev Cancer, 2006. 6(1): p. 11-23.
149. Asahara, T., et al., Isolation of putative progenitor endothelial cells for angiogenesis. Science, 1997. 275(5302): p. 964-967.
150. Komori, T., Regulation of osteoblast differentiation by transcription factors. J Cell Biochem, 2006. 99(5): p. 1233-9.
151. Otto, F., H. Kanegane, and S. Mundlos, Mutations in the RUNX2 gene in patients with cleidocranial dysplasia. Hum Mutat, 2002. 19(3): p. 209-16.
152. Kumagai, K., et al., The extent of degeneration of cruciate ligament is associated with chondrogenic differentiation in patients with osteoarthritis of the knee. Osteoarthritis Cartilage, 2012. 20(11): p. 1258-67.
153. Ng, L.J., et al., SOX9 binds DNA, activates transcription, and coexpresses with type II collagen during chondrogenesis in the mouse. Dev Biol, 1997. 183(1): p. 108-21.
154. Zhao, Q., et al., Parallel expression of Sox9 and Col2a1 in cells undergoing chondrogenesis. Dev Dyn, 1997. 209(4): p. 377-86.
155. Wright, E., et al., The Sry-related gene Sox9 is expressed during chondrogenesis in mouse embryos. Nat Genet, 1995. 9(1): p. 15-20.
156. Murakami, S., et al., Up-regulation of the chondrogenic Sox9 gene by fibroblast growth factors is mediated by the mitogen-activated protein kinase pathway. Proc Natl Acad Sci U S A, 2000. 97(3): p. 1113-8.
157. Chou, Y.H., et al., Intermediate filament reorganization during mitosis is mediated by p34cdc2 phosphorylation of vimentin. Cell, 1990. 62(6): p. 1063-71.
158. Chou, Y.H., et al., The relative roles of specific N- and C-terminal phosphorylation sites in the disassembly of intermediate filament in mitotic BHK-21 cells. J Cell Sci, 1996. 109 ( Pt 4): p. 817-26.
159. Chou, Y.H., et al., Nestin promotes the phosphorylation-dependent disassembly of vimentin intermediate filaments during mitosis. Mol Biol Cell, 2003. 14(4): p. 1468-78.
160. Eliasson, C., et al., Intermediate filament protein partnership in astrocytes. J Biol Chem, 1999. 274(34): p. 23996-4006.
161. Kachinsky, A.M., J.A. Dominov, and J.B. Miller, Intermediate filaments in cardiac myogenesis: nestin in the developing mouse heart. J Histochem Cytochem, 1995. 43(8): p. 843-7.
162. Michalczyk, K. and M. Ziman, Nestin structure and predicted function in cellular cytoskeletal organisation. Histol Histopathol, 2005. 20(2): p. 665-71.
163. Otterbein, L.R., P. Graceffa, and R. Dominguez, The crystal structure of uncomplexed actin in the ADP state. Science, 2001. 293(5530): p. 708-11.
164. Doherty, G.J. and H.T. McMahon, Mediation, modulation, and consequences of membrane-cytoskeleton interactions. Annu Rev Biophys, 2008. 37: p. 65-95.






指導教授 ?口亞紺(Akon Higuchi) 審核日期 2014-6-16
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