博碩士論文 101324033 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:14 、訪客IP:13.59.187.201
姓名 王品毓(Pin-Yu Wang)  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 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
(Proliferation and Differentiation of Amniotic Fluid-Derived Stem Cells on Oligopeptide-Grafted Surface 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
★ 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★ 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
★ 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) 從人類羊水來源的幹細胞是一種多能性幹細胞,因為其具備分化多種譜系的能力,包括代表性的三個胚層。因此,羊水來源幹細胞可能成為一個更適合的幹細胞在再生醫學和組織工程。此外,細胞培養基材的物理特性會影響幹細胞分化的命運。然而,在過去研究報告上沒有探討出最佳的組合以結合物理線索中的彈性及生物線索中的細胞培養基質,以保持幹細胞的多能性和長時間細胞的培養。
因此,在我的研究中,為了探討維持羊水幹細胞的多能性以及如何使羊水幹細胞有效地分化。因為我將羊水來源幹細胞培養於具有不同軟硬度的水凝膠且固定細胞外基質衍生的寡?以提升羊水幹細胞的多能性基因且有效調節細胞的分化能力。一開始,準備polyvinylalcohol-co-itaconic acid (PVA-IA)塗層於細胞培養層上,藉由不同交聯時間已達成控制PVA-IA具有不同的軟硬度,經過活化的程序,便能將細胞外基質衍生的寡?固定在PVA-IA塗層上,最後將羊水幹細胞培養於這些盤子上,以便後續的分析及探索。經過細胞經過數天的培養後,將種於不同條件的羊水幹細胞取其檢測多能性基因:Oct4, Nanog, 和Sox2,和分化基因:Nestin, Sox17, 和Runx2。在最後的檢測結果指出vitronectin來源寡?固定於低彈性的PVA-IA塗層具有較大的潛能以提升羊水幹細胞的多能性;而且在不同軟硬度的PVA-IA塗層能使羊水分化成相似於基材軟硬度的細胞類型,例如:當羊水幹細胞培養於較低彈性的PVA-IA嫁接塗層,會使的羊水幹細胞偏好分化成神經類型細胞;然而,當細胞培養於較高彈性的PVA-IA嫁接途層,將使的細胞分化成成骨幹細胞型態,這項研究結果報導出與先前研究一樣的結果。
因為這項研究中指出,結合培養材料的鋼度以及細胞外基質成份的生物線索可以引導和決定幹細胞多能細和分化的譜系。另一方面,我們進一步發現,不同軟印度和細胞外基質寡?之間的合作將有效地調節羊水幹細胞分化成不同譜系的承諾。
摘要(英) Stem cells derived from human amniotic fluid (AFSCs) are pluripotent fetal cells capable of differentiating into multiple lineages, including representatives of the three embryonic germ layers. Therefore, AFSCs may become a more suitable source of stem cells in regenerative medicine and tissue engineering. However, stem cell characteristics, such as proper differentiation and maintenance of pluripotency, are notregulated only by the stem cells themselves but also by their microenvironment. Furthermore, physical characteristics of cell culture substrate influence the fate of stem cell differentiation. However, there have been no reports from our database study that investigates the optimal elasticity to keep pluripotency of stem cells for a long time and studies the optimal combination of physical cue and biological cue on cell culture substrates. Here I report pluripotent maintenance and differentiation efficiency of AFSCs cultured on cell culture substrates immobilized extracellular matrix-derived oligopeptides, which have different elasticity. We prepared dishes coated with polyvinylalcohol-co-itaconic acid (PVA-IA) films having different elasticity by controlling the crosslinking time in crosslinking solution containing glutaraldehyde, and grafted with several ECM-derived cell-adhesion peptides though N-(3-dimethylaminopropyl)-N’-ethylcarbodiimide hydrochloride(EDC) and N-hydroxysuccinimide (NHS) chemistry in an aqueous solution. qRT-PCR measurements suggest that pluripotent genes, Nanog, Oct4 and Sox2, were kept on PVA-IA hydrogels grafted with oligopeptide derived from vitronectin (oligoVN) and fibronectin (oligoFN) having moderate elasticity around 12-25kPacompared toAFSCs on conventional tissue culture dishes. I found that there is an optimal elasticity of cell culture matrix to promote pluripotency of AFSCs for their culture. Furthermore, early differentiation marker of osteoblasts (Runx2) were found on AFSCs cultured on stiffer PVA-IA hydrogels grafted with oligopeptides derived from Vitronectin (oligoVN) in expansion medium without any induction (differentiation) components, suggesting stiffer culture substrates grafted with oligoVNguide AFSCs into osteoblast lineage, whereas early neural differentiation marker of nestin were more expressed on softer PVA-IA hydrogels grafted with oligoFN in the expansion medium, suggesting softer culture substrates grafted with oligoFN and oligoVN promote differentiation into neural cell lineages. It is suggested that physical cues such as stiffness of culture materials as well as biological cues of extracellular matrix components can guide and decide pluripotency and differentiation lineages of stem cells. On the other hands, I further discoveredthatAFSCs cultured on thecooperation between ECM-ligands and stiffness matrices have been engaged into different lineages commitments.
關鍵字(中) ★ 多能性幹細胞
★ 羊水幹細胞
關鍵字(英) ★ pluripotent stem cell
★ Amniotic Fluid-Derived Stem Cells
論文目次 CHAPTER ONE INTRODUCTION 1
1-1 Stem cell 1
1-1-1 Potency of stem cells 2
1-1-2 Totipotency 2
1-1-3 Pluripotency 2
1-1-4 Multipotency 2
1-2 Souces of stem cells 2
1-2-1 Embryonic stem cells (ESCs) 3
1-2-2 Induced Pluripotent Stem Cells 4
1-2-3 Mesenchymal stem cells (MSCs) 4
1-3 Amniotic fluid-derived stem Cells 6
1-3-2 Amniotic fluid cell type 7
1-3-3 Isolation of amniotic fluid stem cell (AFSCs) 10
1-3-4 Characterization of amniotic fluid stem cell 10
1-3-5 Pluripotency of amniotic fluid stem cells 12
1-4 Niches of stem cells 15
1-4-1 Soluble factors 16
1-4-2 Cell-cell interactions 16
1-4-3 Physical cues affect ex vivo expansion 16
1-4-4 Cell-biomaterials interactions 20
1-5 Extracellular-matrix (ECM) and ECM-mimicking oligopeptides 20
1-5-1 Type and classification of artificial ECMs 23
1-6 The effect of extracellular matrix (ECM) to stem cells 25
1-7 Markers of pluripotent gene 26
1-8 Markers of differentiation lineages of MSCs 28
1-9 Polymerase chain reaction (PCR) 30
1-9-1 Procedure of PCR 30
1-9-2 Reverse transcription polymerase chain reaction (RT-PCR) 32
&Quantitative real time polymerase chain reaction(qRT-PCR) 32
1-9-3 Procedure of RT-PCR 33
CHAPTER TWO MATERIALS AND METHODS 35
2-1 Materials 35
2-1-1 Cultured medium 35
2-1-2 Serum 35
2-1-3 Antibiotics 35
2-1-4 Growth factor 35
2-1-5 PVA-IA film 35
2-1-6 ECM proteins, ECM-derived peptides and CellStart 36
2-1-8 RNA extraction 36
2-1-9 Reverse transcriptase (RT) 36
2-1-10 Real-time PCR (qPCR) 36
2-1-11 Probes for qPCR 37
2-2 Methods and Analysis 37
2-2-1 PVA-IA film preparation 37
2-2-2 Preparation of PVA-IA surfaces grafted with ECM and ECM-derived oligopeptides 39
2-2-3 Elasticity measurement of PVA-IA 40
2-2-4 XPS analysis of dish surface 41
2-2-5 Phosphate buffer saline (PBS) preparation 41
2-2-6 Preparation of FGF-2(β-FGF) protein stock solution 42
2-2-7 Preparation of cell culture medium 42
2-2-8 Cell cultivation 42
2-2-9 Cell density measurement 43
2-2-10 Isolation of total RNA 44
2-2-11 Reverse Transcription of mRNA into cDNA 44
2-2-12 Quantitative real time polymerase chain reaction 45
2-2-13 Imunofluorescence assay 46
CHAPTER THREE RESULTS 49
3-1 The elasticity of the PVA-IA films 49
3-2 XPS measurements of the PVA-IA films (12.2kPa &25.3kPa) grafted with ECM and ECM-derived peptides 52
3-3 Proliferation of AFSCs cultured on soft and stiff PVA-IA films grafted with ECM/ECM-derived peptides 61
3-4 Up-regulating pluripotency of AFSCs cultured on soft and stiff PVA-IA films grafted with and ECM and ECM-derived peptides 70
3-5 Directing AFSC fates on PVA-IA films by controlling matrix elasticity and extracellular adhesion ligand 76
CHAPTER FOUR DISSCUSION 87
CHAPTER FIVE CONCLUSION 91




INDEX OF FIGURES
Figure 1-1 Stem cell pathways [3]. 1
Figure 1-2 Embryonic stem cells are isolation from the inner cell mass of blastocyst [19]. 3
Figure 1-3 Morphology of MSCs derived from amniotic stem fluid. 5
Figure 1-4 MSC differentiation [28]. 5
Figure 1-5 The time line of pregnant period. 7
Figure 1-6 Mature primary colonies of amniotic fluid colony cells after 14-16 days in culture:(a) F-type, (b) AF-type, (c) E-type [30]. 8
Figure 1-7 Extraction of amniotic fluid [37]. 11
Figure 1-8 Surface marker expression of hAFSCs [10]. 12
Figure 1-9 The microenvironment and niches of stem cells and their regulation [26]. 15
Figure 1-10 Tissue elasticity and differentiation of native MSCs [63]. 18
Figure 1-11 Substrate stiffness influenced adhesion structures and dynamics [66]. 19
Figure 1-12 Actin cytoskeleton on cell-on-cell layering [70]. 19
Figure 1-13 PCR mechanism [154]. 31
Figure1-14 Flow chart of the technical steps in qRT-PCR [160]. 33
Figure1-15 Process of qRT-PCR. 34
Figure 2-1 The process of cross linked PVA-IA coating dish grafted with nano-segment. 38
Figure 2-2 Controlling the stiffness of PVA-IA films. 38
Figure 2-3 ECMs and ECM-derived peptides used in this experiment. 39
Figure 2-4 Two major types of immunofluorescence staining. 48
Figure 3-1 The storage modulus of crosslinking PVA-IA films at 1Hz angular frequency. 50
Figure 3-2 The storage modulus of crosslinking PVA-IA films at different angular frequency. 51
Figure 3-3 The storage modulus of crosslinking PVA-IA films at 1Hz of angular frequency. 52
Figure 3-4 The elemental analysis of PVA-IA films (12.2kPa) grafted with ECM and ECM-derived peptides by X-ray photoelectron spectroscopy. 54
Figure 3-5 The elemental analysis of PVA-IA films grafted with ECM and ECM-derived peptides and unmodified PVA-IA film (12.2kPa) by X-ray elemental photoelectron spectroscopy. 55
Figure 3-6 The elemental analysis of PVA-IA films (25.3kPa) grafted with ECM and ECM-derived peptides by X-ray photoelectron spectroscopy. 56
Figure 3-7 The analysis of PVA-IA films grafted with ECM, ECM-derived peptides and unmodified PVA-IA film (25.3kPa) by X-ray photoelectron spectroscopy. 57
Figure 3-8 The elemental analysis of TCPS by X-ray photoelectron spectroscopy. 58
Figure 3-9 O/C ratio of TCPS, PVA-IA films grafted with ECM and ECM-derived peptides and unmodified PVA-IA film. 59
Figure 3-10 N/C ratio of TCPS, PVA-IA films grafted with ECM and ECM-derived peptides and unmodified PVA-IA film. 59
Figure 3-11 The morphologies of AFSCs cultured on PVA-IA films and PVA-IA films (10.8 and 14.6kPa) grafting with COL and COL-derived peptides at day 7 culture. 63
Figure 3-12 The morphologies of AFSCs cultured on PVA-IA films (10.8 and 14.6kPa) grafting with FN, VN and their derived peptides at day 7 culture. 64
Figure 3-13 The morphologies of AFSCs cultured on PVA-IA films and PVA-IA films (12.2 and 25.3kPa) grafting with COL and COL-derived peptides at day 7 culture. 65
Figure 3-14 The morphologies of AFSCs cultured on PVA-IA films (12.2 and 25.3kPa) grafting with FN, VN and their derived peptides at day 7 culture. 66
Figure 3-15 The morphologies of AFSCs cultured on CellStart and TCPS at day 7 culture. 67
Figure 3-16 Growth curve of AFSCs cultured on soft (12.2kPa) PVA-IA films grafted with ECM and ECM-derived peptides at passage 4. 68
Figure 3-17 Growth curve of AFSCs cultured on stiff (25.3kPa) PVA-IA films grafted with ECM and ECM-derived peptides at passage 4. 68
Figure 3-18 Doubling time of AFSCs cultured on PVA-IA films (12.2kPa and 25.3kPa) grafted with ECM and ECM-derived peptides at passage 4. 69
Figure 3-19 Pluripotent genes expression of AFSCs on soft and stiff (12.2 &25.3kPa) PVA-IA films immobilized with ECM and ECM-derived peptides were measured by real-time PCR. 73
Figure 3-20 Pluripotent gene expression of AFSCs on the soft (12.2kPa) PVA-IA films immobilized with ECM and ECM-derived peptides. 74
Figure 3-21 Pluripotent gene expression of AFSCs on the stiff (25.3kPa) PVA-IA films immobilized with ECM and ECM-derived peptides. 75
Figure 3-22 Lineage-specific gene expression of AFSCs cultured on the soft and stiff (12.2 & 25.3 kPa) PVA-IA films immobilized with ECM and ECM-derived peptides by qPCR. 79
Figure 3-23 Immunofluorescences staining for Hochest (blue), Sox2 (green) and β3-tubulin (red) in AFSCs after 7 days of culture on different dishes. 81
Figure 3-24 Immunofluorescences staining for Hochest (blue), Sox2 (green) and β3-tubulin (red) in AFSCs after 7 days of culture on different dishes. 82
Figure 3-25 Immunofluorescences staining for Hochest (blue), Oct4 (red) and α-SMA (green) in AFSCs after 7 days of culture on different dishes. 83
Figure 3-26 Immunofluorescences staining for Hochest (blue), Oct4 (red) and α-SMA (green) in AFSCs after 7 days of culture on different dishes. 84
Figure 3-27 Immunofluorescences staining for Hochest (blue), SSEA4 (green) and AFP (red) in AFSCs after 7 days of culture on different dishes. 85
Figure 3-28 Immunofluorescences staining for Hochest (blue), SSEA4 (green) and AFP (red) in AFSCs after 7 days of culture on different dishes. 86
Figure 4-1 Pluripotency of AFSCs cultured on soft and stiff PVA-IA films (12.2 & 25.3kPa) immobilized with ECM and ECM-derived peptides. 89
Figure 4-2 The correlation between pluripotency and specific lineages of differentiation of AFSCs cultured on soft and stiff PVA-IA films (12.2 & 25.3kPa) immobilized with ECM and ECM-derived peptides. 90
參考文獻 1. Ulloa-Montoya, F., C.M. Verfaillie, and W.S. Hu, Culture systems for pluripotent stem cells. Journal of Bioscience and Bioengineering, 2005. 100(1): p. 12-27.
2. Verfaillie, C.M., Pluripotent stem cells. Transfusion Clinique et Biologique, 2009.
3. On line resources : Cell signal.
4. On line resources︰Wikipimedia.
5. Scholer, H.R., The Potential of Stem Cells: An Inventory. 2007, in Nikolaus Knoepffler, Dagmar Schipanski, and Stefan Lorenz Sorgner. Humanbiotechnology as Social Challenge: Ashgate Publishing, Ltd. 28.
6. Reya, T., et al., Stem cells, cancer, and cancer stem cells. Nature, 2001. 414(6859): p. 105-111.
7. Fauza, D., Amniotic fluid and placental stem cells. Best Pract Res Clin Obstet Gynaecol, 2004. 18(6): p. 877-91.
8. Hengstschlager, M., stem cell in amniotic fluid J Reproduktion Med Endocrinol, 2005.
9. Kolambkar, Y.M., et al., Chondrogenic differentiation of amniotic fluid-derived stem cells. Journal of Molecular Histology, 2007. 38(5): p. 405-413.
10. De Coppi, P., et al., Isolation of amniotic stem cell lines with potential for therapy. Nature Biotechnology, 2007. 25(1): p. 100-106.
11. Zheng, Y.B., et al., Characterization and hepatogenic differentiation of mesenchymal stem cells from human amniotic fluid and human bone marrow: A comparative study. Cell Biology International, 2008. 32(11): p. 1439-1448.
12. Tsai, M.S., et al., Functional network analysis of the transcriptomes of mesenchymal stem cells derived from amniotic fluid, amniotic membrane, cord blood, and bone marrow. Stem Cells, 2007. 25(10): p. 2511-23.
13. Kim, J., et al., Human amniotic fluid-derived stem cells have characteristics of multipotent stem cells. Cell Proliferation, 2007. 40(1): p. 75-90.
14. Battula, V.L., et al., Human placenta and bone marrow derived MSC cultured in serum-free, b-FGF-containing medium express cell surface frizzled-9 and SSEA-4 and give rise to multilineage differentiation. Differentiation, 2007. 75(4): p. 279-91.
15. Tsai, M.S., et al., Clonal amniotic fluid-derived stem cells express characteristics of both mesenchymal and neural stem cells. Biology of Reproduction, 2006. 74(3): p. 545-551.
16. Poloni, A., et al., Characterization and expansion of mesenchymal progenitor cells from first-trimester chorionic villi of human placenta. Cytotherapy, 2008. 10(7): p. 690-697.
17. Gucciardo, L., et al., Fetal mesenchymal stem cells: isolation, properties and potential use in perinatology and regenerative medicine. BJOG, 2009. 116(2): p. 166-72.
18. Klimanskaya, I., et al., Human embryonic stem cells derived without feeder cells. Lancet, 2005. 365(9471): p. 1636-1641.
19. Yabut, O. and H.S. Bernstein, The promise of human embryonic stem cells in aging-associated diseases. Aging (Albany NY), 2011. 3(5): p. 494-508.
20. 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.
21. Okita, K., T. Ichisaka, and S. Yamanaka, Generation of germline-competent induced pluripotent stem cells. Nature, 2007. 448(7151): p. 313-U1.
22. Takahashi, K. and S. Yamanaka, Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell, 2006. 126(4): p. 663-676.
23. Yu, J.Y., et al., Induced pluripotent stem cell lines derived from human somatic cells. Science, 2007. 318(5858): p. 1917-1920.
24. Lin, S.L., et al., Mir-302 reprograms human skin cancer cells into a pluripotent ES-cell-like state. Rna-a Publication of the Rna Society, 2008. 14(10): p. 2115-2124.
25. Zhou, H.Y., et al., Generation of Induced Pluripotent Stem Cells Using Recombinant Proteins (vol 4, pg 381, 2009). Cell Stem Cell, 2009. 4(6): p. 581-581.
26. Higuchi, A., et al., Biomimetic Cell Culture Proteins as Extracellular Matrices for Stem Cell Differentiation. Chemical Reviews, 2012. 112(8): p. 4507-4540.
27. Brighton, C.T. and R.M. Hunt, Early Histological and Ultrastructural-Changes in Medullary Fracture Callus. Journal of Bone and Joint Surgery-American Volume, 1991. 73A(6): p. 832-847.
28. MSC differentiation. June 2001, Trends in Molecular Medicine. p. V.7 No. 6
29. Chamberlain, G., et al., Concise review: mesenchymal stem cells: their phenotype, differentiation capacity, immunological features, and potential for homing. Stem Cells, 2007. 25(11): p. 2739-49.
30. GOSDEN, C.M., AMNIOTIC FLUID CELL TYPES AND CULTURE. British Medical Bulletin, 1983. 39(4): p. 348-354.
31. Hoehn, H. and D. Salk, Morphological and biochemical heterogeneity of amniotic fluid cells in culture. Methods Cell Biol., 1982. 26: p. 11-34.
32. 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.
33. Jiang, Y.H., et al., Pluripotency of mesenchymal stem cells derived from adult marrow. Nature, 2002. 418(6893): p. 41-49.
34. Hoehn, H. and D. Salk, Chapter 2. Morphological and Biochemical Heterogeneity of Amniotic Fluid Cells in Culture, in Methods in Cell Biology, A.L. Samuel and J.D. Gretchen, Editors. 1982, Academic Press. p. 11-34.
35. Perrone, L., et al., Postnatal weight change is influenced by mother-newborn pair leptin levels. Nutrition Research, 2000. 20(11): p. 1531-1536.
36. Zambotti, F., et al., Monoamine metabolites and related compounds in human amniotic fluid: Assay by gas chromatography and gas chromatography-mass spectrometry. Clinica Chimica Acta, 1975. 61(3): p. 247-256.
37. On line resources:.
38. Barry, F.P., et al., The monoclonal antibody SH-2, raised against human mesenchymal stem cells, recognizes an epitope on endoglin (CD105). Biochemical and Biophysical Research Communications, 1999. 265(1): p. 134-139.
39. Barry, F., et al., The SH-3 and SH-4 antibodies recognize distinct epitopes on CD73 from human mesenchymal stem cells. Biochemical and Biophysical Research Communications, 2001. 289(2): p. 519-524.
40. Baksh, D., L. Song, and R.S. Tuan, Adult mesenchymal stem cells: characterization, differentiation, and application in cell and gene therapy. Journal of Cellular and Molecular Medicine, 2004. 8(3): p. 301-316.
41. Ririe, K.M., R.P. Rasmussen, and C.T. Wittwer, Product differentiation by analysis of DNA melting curves during the polymerase chain reaction. Analytical Biochemistry, 1997. 245(2): p. 154-160.
42. Shamblott, M.J., et al., Human embryonic germ cell derivatives express a broad range of developmentally distinct markers and proliferate extensively in vitro. Proceedings of the National Academy of Sciences of the United States of America, 2001. 98(1): p. 113-118.
43. Pittenger, M.F., et al., Multilineage potential of adult human mesenchymal stem cells. Science, 1999. 284(5411): p. 143-147.
44. Zuk, P.A., et al., Human adipose tissue is a source of multipotent stem cells. Molecular Biology of the Cell, 2002. 13(12): p. 4279-4295.
45. Kubista, M., et al., Brca1 regulates in vitro differentiation of mammary epithelial cells. Oncogene, 2002. 21(31): p. 4747-4756.
46. Soucek, T., et al., A role of the tuberous sclerosis gene-2 product during neuronal differentiation. Oncogene, 1998. 16(17): p. 2197-2204.
47. Prusa, A.R., et al., Neurogenic cells in human amniotic fluid. American Journal of Obstetrics and Gynecology, 2004. 191(1): p. 309-314.
48. Ter Brugge, P.J. and J.A. Jansen, In vitro osteogenic differentiation of rat bone marrow cells subcultured with and without dexamethasone. Tissue Engineering, 2002. 8(2): p. 321-331.
49. Cooper, G.M., The Cell: A Molecular Approach. 2000-2009.
50. Gilbert, P.M., et al., Substrate Elasticity Regulates Skeletal Muscle Stem Cell Self-Renewal in Culture. Science, 2010. 329(5995): p. 1078-1081.
51. Georges, P.C., et al., Matrices with compliance comparable to that of brain tissue select neuronal over glial growth in mixed cortical cultures. Biophysical Journal, 2006. 90(8): p. 3012-3018.
52. Flanagan, L.A., et al., Neurite branching on deformable substrates. Neuroreport, 2002. 13(18): p. 2411-2415.
53. Hofstetter, C.P., et al., Marrow stromal cells form guiding strands in the injured spinal cord and promote recovery. Proceedings of the National Academy of Sciences of the United States of America, 2002. 99(4): p. 2199-2204.
54. Kondo, T., et al., Sonic hedgehog and retinoic acid synergistically promote sensory fate specification from bone marrow-derived pluripotent stem cells. Proceedings of the National Academy of Sciences of the United States of America, 2005. 102(13): p. 4789-4794.
55. Engler, A.J., et al., Myotubes differentiate optimally on substrates with tissue-like stiffness: pathological implications for soft or stiff microenvironments. Journal of Cell Biology, 2004. 166(6): p. 877-887.
56. Ferrari, G., et al., Muscle regeneration by bone marrow derived myogenic progenitors. Science, 1998. 279(5356): p. 1528-1530.
57. Andrades, J.A., et al., Selection and amplification of a bone marrow cell population and its induction to the chondro-osteogenic lineage by rhOP-1: an in vitro and in vivo study. International Journal of Developmental Biology, 2001. 45(4): p. 689-693.
58. Holmbeck, K., et al., MT1-MMP-deficient mice develop dwarfism, osteopenia, arthritis, and connective tissue disease due to inadequate collagen turnover. Cell, 1999. 99(1): p. 81-92.
59. Morinobu, M., et al., Osteopontin expression in osteoblasts and osteocytes during bone formation under mechanical stress in the calvarial suture in vivo. Journal of Bone and Mineral Research, 2003. 18(9): p. 1706-1715.
60. Deng, J., et al., Mesenchymal stem cells spontaneously express neural proteins in culture and are neurogenic after transplantation. Stem Cells, 2006. 24(4): p. 1054-1064.
61. McBeath, R., et al., Cell shape, cytoskeletal tension, and RhoA regulate stem cell lineage commitment. Developmental Cell, 2004. 6(4): p. 483-495.
62. LaIuppa, J.A., et al., Culture materials affect ex vivo expansion of hematopoietic progenitor cells. Journal of Biomedical Materials Research, 1997. 36(3): p. 347-359.
63. Engler, A.J., et al., Matrix elasticity directs stem cell lineage specification. Cell, 2006. 126(4): p. 677-689.
64. Seraj, M.J., et al., Functional evidence for a novel human breast carcinoma metastasis suppressor, BRMS1, encoded at chromosome 11q13. Cancer Research, 2000. 60(11): p. 2764-2769.
65. Discher, D.E., P. Janmey, and Y.L. Wang, Tissue cells feel and respond to the stiffness of their substrate. Science, 2005. 310(5751): p. 1139-1143.
66. Pelham, R.J. and Y.-l. Wang, Cell locomotion and focal adhesions are regulated by substrate?flexibility. Proceedings of the National Academy of Sciences, 1997. 94(25): p. 13661-13665.
67. Engler, A., et al., Substrate compliance versus ligand density in cell on gel responses. Biophysical Journal, 2004. 86(1 Pt 1): p. 617-28.
68. Georges, P.C. and P.A. Janmey, Cell type-specific response to growth on soft materials. J Appl Physiol (1985), 2005. 98(4): p. 1547-53.
69. Engler, A.J., et al., Myotubes differentiate optimally on substrates with tissue-like stiffness: pathological implications for soft or stiff microenvironments. Journal of Cell Biology, 2004. 166(6): p. 877-87.
70. Discher, D.E., P. Janmey, and Y.L. Wang, Tissue cells feel and respond to the stiffness of their substrate. Science, 2005. 310(5751): p. 1139-43.
71. Lu, Z., et al., Collagen Type II Enhances Chondrogenesis in Adipose Tissue-Derived Stem Cells by Affecting Cell Shape. Tissue Engineering Part A, 2010. 16(1): p. 81-90.
72. 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.
73. Jiang, X.S., et al., Surface-immobilization of adhesion peptides on substrate for ex vivo expansion of cryopreserved umbilical cord blood CD34(+) cells. Biomaterials, 2006. 27(13): p. 2723-2732.
74. Gelain, F., et al., Designer Self-Assembling Peptide Nanofiber Scaffolds for Adult Mouse Neural Stem Cell 3-Dimensional Cultures. Plos One, 2006. 1(2).
75. Frontiers in tissue engineering. 1998.
76. Rosso, F., et al., From cell-ECM interactions to tissue engineering. Journal of Cellular Physiology, 2004. 199(2): p. 174-80.
77. Langer, R. and J. Vacanti, Tissue engineering. Science, 1993. 260(5110): p. 920-926.
78. R.Barbucci, Integrated Biomaterials Science. 2002.
79. Putnam, A.J. and D.J. Mooney, Tissue engineering using synthetic extracellular matrices. Nature Medicine, 1996. 2(7): p. 824-826.
80. Daley, W.P., S.B. Peters, and M. Larsen, Extracellular matrix dynamics in development and regenerative medicine. Journal of Cell Science, 2008. 121(Pt 3): p. 255-64.
81. Rozario, T. and D.W. DeSimone, The extracellular matrix in development and morphogenesis: a dynamic view. Developmental Biology, 2010. 341(1): p. 126-40.
82. 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.
83. Lee, C.H., A. Singla, and Y. Lee, Biomedical applications of collagen. International Journal of Pharmaceutics, 2001. 221(1-2): p. 1-22.
84. Pankov, R. and K.M. Yamada, Fibronectin at a glance. Journal of Cell Science, 2002. 115(20): p. 3861-3863.
85. Mao, Y. and J.E. Schwarzbauer, Fibronectin fibrillogenesis, a cell-mediated matrix assembly process. Matrix Biology, 2005. 24(6): p. 389-399.
86. resources:, O.l. http://www.sigmaaldrich.com/catalog/product/sigma/V8379?lang=enRion=TW.
87. Haque, A., et al., The effect of recombinant E-cadherin substratum on the differentiation of endoderm-derived hepatocyte-like cells from embryonic stem cells. Biomaterials, 2011. 32(8): p. 2032-42.
88. Kaur, G., et al., The promotion of osteoblastic differentiation of rat bone marrow stromal cells by a polyvalent plant mosaic virus. Biomaterials, 2008. 29(30): p. 4074-81.
89. Yue, X.S., et al., A fusion protein N-cadherin-Fc as an artificial extracellular matrix surface for maintenance of stem cell features. Biomaterials, 2010. 31(20): p. 5287-96.
90. Shi, C., et al., Stem-cell-capturing collagen scaffold promotes cardiac tissue regeneration. Biomaterials, 2011. 32(10): p. 2508-15.
91. Lee, H.J., et al., Enhanced chondrogenesis of mesenchymal stem cells in collagen mimetic peptide-mediated microenvironment. Tissue Eng Part A, 2008. 14(11): p. 1843-51.
92. You, M., et al., Chondrogenic differentiation of human bone marrow mesenchymal stem cells on polyhydroxyalkanoate (PHA) scaffolds coated with PHA granule binding protein PhaP fused with RGD peptide. Biomaterials, 2011. 32(9): p. 2305-13.
93. Hennessy, K.M., et al., The effect of collagen I mimetic peptides on mesenchymal stem cell adhesion and differentiation, and on bone formation at hydroxyapatite surfaces. Biomaterials, 2009. 30(10): p. 1898-909.
94. Yang, F., et al., The effect of incorporating RGD adhesive peptide in polyethylene glycol diacrylate hydrogel on osteogenesis of bone marrow stromal cells. Biomaterials, 2005. 26(30): p. 5991-8.
95. Nguyen, L.H., et al., Unique biomaterial compositions direct bone marrow stem cells into specific chondrocytic phenotypes corresponding to the various zones of articular cartilage. Biomaterials, 2011. 32(5): p. 1327-38.
96. Betre, H., et al., Chondrocytic differentiation of human adipose-derived adult stem cells in elastin-like polypeptide. Biomaterials, 2006. 27(1): p. 91-9.
97. Meinel, L., et al., Engineering bone-like tissue in vitro using human bone marrow stem cells and silk scaffolds. Journal of Biomedical Materials Research Part A, 2004. 71(1): p. 25-34.
98. 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. 2962-9.
99. Wojtowicz, A.M., et al., Coating of biomaterial scaffolds with the collagen-mimetic peptide GFOGER for bone defect repair. Biomaterials, 2010. 31(9): p. 2574-82.
100. Cooke, M.J., et al., Neural differentiation regulated by biomimetic surfaces presenting motifs of extracellular matrix proteins. Journal of Biomedical Materials Research Part A, 2010. 93(3): p. 824-32.
101. Gelain, F., et al., Designer Self-Assembling Peptide Nanofiber Scaffolds for Adult Mouse Neural Stem Cell 3-Dimensional Cultures. Plos One, 2006. 1(1): p. e119.
102. Anderson, J.M., et al., Osteogenic differentiation of human mesenchymal stem cells directed by extracellular matrix-mimicking ligands in a biomimetic self-assembled peptide amphiphile nanomatrix. Biomacromolecules, 2009. 10(10): p. 2935-44.
103. Bhatnagar, R.S., J.J. Qian, and C.A. Gough, The role in cell binding of a beta-bend within the triple helical region in collagen alpha 1 (I) chain: structural and biological evidence for conformational tautomerism on fiber surface. J Biomol Struct Dyn, 1997. 14(5): p. 547-60.
104. Jiang, X.S., et al., Surface-immobilization of adhesion peptides on substrate for ex vivo expansion of cryopreserved umbilical cord blood CD34+ cells. Biomaterials, 2006. 27(13): p. 2723-32.
105. Higuchi, A., et al., Polymeric Materials for Ex vivo Expansion of Hematopoietic Progenitor and Stem Cells. Polymer Reviews, 2009. 49(3): p. 181-200.
106. 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. 606-10.
107. 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. 30-3.
108. Suzuki, S., et al., Complete amino acid sequence of human vitronectin deduced from cDNA. Similarity of cell attachment sites in vitronectin and fibronectin. Embo Journal, 1985. 4(10): p. 2519-24.
109. Oldberg, A., A. Franzen, and D. Heinegard, The primary structure of a cell-binding bone sialoprotein. Journal of Biological Chemistry, 1988. 263(36): p. 19430-2.
110. Park, I.S., et al., The correlation between human adipose-derived stem cells differentiation and cell adhesion mechanism. Biomaterials, 2009. 30(36): p. 6835-43.
111. Rosso, F., et al., Smart materials as scaffolds for tissue engineering. Journal of Cellular Physiology, 2005. 203(3): p. 465-470.
112. 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.
113. 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.
114. Scadden, D.T., The stem-cell niche as an entity of action. Nature, 2006. 441(7097): p. 1075-1079.
115. Nilsson, S.K., et al., Osteopontin, a key component of the hematopoietic stem cell niche and regulator of primitive hematopoietic progenitor cells. Blood, 2005. 106(4): p. 1232-1239.
116. Moore, K.A. and I.R. Lemischka, Stem cells and their niches. Science, 2006. 311(5769): p. 1880-1885.
117. Schofield, R., The relationship between the spleen colony-forming cell and the haemopoietic stem cell. Blood Cells, 1978. 4(1-2): p. 7-25.
118. Li, L.H. and T. Xie, Stem cell niche: Structure and function. Annual Review of Cell and Developmental Biology, 2005. 21: p. 605-631.
119. Jensen, U.B., S. Lowell, and F.M. Watt, The spatial relationship between stem cells and their progeny in the basal layer of human epidermis: a new view based on whole-mount labelling and lineage analysis. Development, 1999. 126(11): p. 2409-2418.
120. 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.
121. Nuttelman, C.R., M.C. Tripodi, and K.S. Anseth, Synthetic hydrogel niches that promote hMSC viability. Matrix Biology, 2005. 24(3): p. 208-218.
122. Feng, Q., et al., Expansion of engrafting human hematopoietic stem/progenitor cells in three-dimensional scaffolds with surface-immobilized fibronectin. Journal of Biomedical Materials Research Part A, 2006. 78A(4): p. 781-791.
123. Gerecht, S., et al., Hyaluronic acid hydrogen for controlled self-renewal and differentiation of human embryonic stem cells. Proceedings of the National Academy of Sciences of the United States of America, 2007. 104(27): p. 11298-11303.
124. Chang, C.-F., et al., Three-dimensional collagen fiber remodeling by mesenchymal stem cells requires the integrin–matrix interaction. Journal of Biomedical Materials Research Part A, 2007. 80A(2): p. 466-474.
125. Donovan, P.J. and J. Gearhart, The end of the beginning for pluripotent stem cells. Nature, 2001. 414(6859): p. 92-97.
126. 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.
127. Scholer, H.R., et al., Oct-4 - a Germline-Specific Transcription Factor Mapping to the Mouse T-Complex. Embo Journal, 1990. 9(7): p. 2185-2195.
128. Scholer, H.R., et al., New Type of Pou Domain in Germ Line-Specific Protein Oct-4. Nature, 1990. 344(6265): p. 435-439.
129. Avilion, A.A., et al., Multipotent cell lineages in early mouse development depend on SOX2 function. Genes & Development, 2003. 17(1): p. 126-140.
130. Chambers, I., et al., Functional expression cloning of Nanog, a pluripotency sustaining factor in embryonic stem cells. Cell, 2003. 113(5): p. 643-655.
131. Mitsui, K., et al., The homeoprotein Nanog is required for maintenance of pluripotency in mouse epiblast and ES cells. Cell, 2003. 113(5): p. 631-642.
132. 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.
133. Darr, H., Y. Mayshar, and N. Benvenisty, Overexpression of NANOG in human ES cells enables feeder-free growth while inducing primitive ectoderm features. Development, 2006. 133(6): p. 1193-1201.
134. Zaehres, H., et al., High-efficiency RNA interference in human embryonic stem cells. Stem Cells, 2005. 23(3): p. 299-305.
135. Chambers, I. and S.R. Tomlinson, The transcriptional foundation of pluripotency. Development, 2009. 136(14): p. 2311-2322.
136. Masui, S., et al., Pluripotency governed by Sox2 via regulation of Oct3/4 expression in mouse embryonic stem cells. Nature Cell Biology, 2007. 9(6): p. 625-U26.
137. 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 Research, 1992. 20(17): p. 4613-4620.
138. Boyer, L.A., et al., Core transcriptional regulatory circuitry in human embryonic stem cells. Cell, 2005. 122(6): p. 947-956.
139. Looijenga, L.H.J., et al., POU5F1 (OCT3/4) identifies cells with pluripotent potential in human germ cell tumors. Cancer Research, 2003. 63(9): p. 2244-2250.
140. Rodda, D.J., et al., Transcriptional regulation of Nanog by Oct4 and Sox2. Journal of Biological Chemistry, 2005. 280(26): p. 24731-24737.
141. Otto, F., et al., Cbfa1, a candidate gene for cleidocranial dysplasia syndrome, is essential for osteoblast differentiation and bone development. Cell, 1997. 89(5): p. 765-771.
142. Komori, T., Regulation of osteoblast differentiation by transcription factors. Journal of Cellular Biochemistry, 2006. 99(5): p. 1233-1239.
143. Otto, F., H. Kanegane, and S. Mundlos, Mutations in the RUNX2 gene in patients with clelidocranial dysplasia. Human Mutation, 2002. 19(3): p. 209-216.
144. Aubin, J.E., et al., Intermediate filaments of the vimentin-type and the cytokeratin-type are distributed differently during mitosis. Exp Cell Res, 1980. 129(1): p. 149-65.
145. Chou, Y.H., et al., Intermediate Filament Reorganization during Mitosis Is Mediated by P34cdc2 Phosphorylation of Vimentin. Cell, 1990. 62(6): p. 1063-1071.
146. 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. Journal of Cell Science, 1996. 109: p. 817-826.
147. Chou, Y.H., et al., Nestin promotes the phosphorylation-dependent disassembly of vimentin intermediate filaments during mitosis. Molecular Biology of the Cell, 2003. 14(4): p. 1468-78.
148. Eliasson, C., et al., Intermediate filament protein partnership in astrocytes. Journal of Biological Chemistry, 1999. 274(34): p. 23996-24006.
149. Kachinsky, A.M., J.A. Dominov, and J.B. Miller, Intermediate filaments in cardiac myogenesis: nestin in the developing mouse heart. The journal of histochemistry and cytochemistry : official journal of the Histochemistry Society, 1995. 43(8): p. 843-847.
150. Michalczyk, K. and M. Ziman, Nestin structure and predicted function in cellular cytoskeletal organisation. Histology and Histopathology, 2005. 20(2): p. 665-671.
151. Seguin, C.A., et al., Establishment of endoderm progenitors by SOX transcription factor expression in human embryonic stem cells. Cell Stem Cell, 2008. 3(2): p. 182-195.
152. D. John O′Connor, B.A.S., Roger St. C. Smart, Surface analysis methods in materials science, Springer-Verlag.
153. Chadwick, K., et al., Cytokines and BMP-4 promote hematopoietic differentiation of human embryonic stem cells. Blood, 2003. 102(3): p. 906-915.
154. Chien, A., D.B. Edgar, and J.M. Trela, Deoxyribonucleic acid polymerase from the extreme thermophile Thermus aquaticus. J Bacteriol, 1976. 127(3): p. 1550-7.
155. Freeman, W.M., S.J. Walker, and K.E. Vrana, Quantitative RT-PCR: Pitfalls and potential. Biotechniques, 1999. 26(1): p. 112-+.
156. Lawyer, F.C., et al., High-level expression, purification, and enzymatic characterization of full-length Thermus aquaticus DNA polymerase and a truncated form deficient in 5′ to 3′ exonuclease activity. PCR Methods Appl, 1993. 2(4): p. 275-87.
157. Simmons, P.J. and B. Torokstorb, Identification of Stromal Cell Precursors in Human Bone-Marrow by a Novel Monoclonal-Antibody, Stro-1. Blood, 1991. 78(1): p. 55-62.
158. Gilliland, G., et al., Analysis of cytokine mRNA and DNA: detection and quantitation by competitive polymerase chain reaction. Proc Natl Acad Sci U S A, 1990. 87(7): p. 2725-9.
159. Rappolee, D.A., et al., Wound macrophages express TGF-alpha and other growth factors in vivo: analysis by mRNA phenotyping. Science, 1988. 241(4866): p. 708-12.
160. Becker-Andre, M. and K. Hahlbrock, Absolute mRNA quantification using the polymerase chain reaction (PCR). A novel approach by a PCR aided transcript titration assay (PATTY). Nucleic Acids Research, 1989. 17(22): p. 9437-46.
161. Zhang, S.C., et al., In vitro differentiation of transplantable neural precursors from human embryonic stem cells. Nature Biotechnology, 2001. 19(12): p. 1129-1133.
162. Cheng, S., et al., Effective Amplification of Long Targets from Cloned Inserts and Human Genomic DNA. Proceedings of the National Academy of Sciences of the United States of America, 1994. 91(12): p. 5695-5699.
163. Holland, P.M., et al., Detection of Specific Polymerase Chain-Reaction Product by Utilizing the 5′-]3′ Exonuclease Activity of Thermus-Aquaticus DNA-Polymerase. Proceedings of the National Academy of Sciences of the United States of America, 1991. 88(16): p. 7276-7280.
164. Witkowska-Zimny, M., et al., Effect of substrate stiffness on differentiation of umbilical cord stem cells. Acta Biochim Pol, 2012. 59(2): p. 261-4.
165. 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.
166. Higuchi, A., et al., Osteoblast differentiation of amniotic fluid-derived stem cells irradiated with visible light. Tissue Eng Part A, 2011. 17(21-22): p. 2593-602.
167. Huebsch, N., et al., Harnessing traction-mediated manipulation of the cell/matrix interface to control stem-cell fate. Nature Materials, 2010. 9(6): p. 518-526.
168. Benoit, D.S.W., et al., Small functional groups for controlled differentiation of hydrogel-encapsulated human mesenchymal stem cells. Nature Materials, 2008. 7(10): p. 816-823.
169. Zemel, A., et al., Optimal matrix rigidity for stress-fibre polarization in stem cells. Nature Physics, 2010. 6(6): p. 468-473.
170. Trappmann, B., et al., Extracellular-matrix tethering regulates stem-cell fate. Nature Materials, 2012. 11(7): p. 642-649.
171. Lee, J., et al., Directing stem cell fate on hydrogel substrates by controlling cell geometry, matrix mechanics and adhesion ligand composition. Biomaterials, 2013. 34(33): p. 8140-8.
172. Higuchi, A., et al., Physical Cues of Biomaterials Guide Stem Cell Differentiation Fate. Chemical Reviews, 2013. 113(5): p. 3297-3328.
173. Zouani, O.F., et al., Effect of BMP-2 from matrices of different stiffnesses for the modulation of stem cell fate. Biomaterials, 2013. 34(9): p. 2157-66.

指導教授 ?口亞紺(Akon Higuchi) 審核日期 2014-6-27
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