博碩士論文 105324062 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:46 、訪客IP:18.119.107.96
姓名 曾也家(Yeh-Chia Tseng)  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 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
(Molecular Design of Nanosegments on Cell Culture Biomaterials for Proliferation of Human Embryonic Stem Cells)
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
★ 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
檔案 [Endnote RIS 格式]    [Bibtex 格式]    [相關文章]   [文章引用]   [完整記錄]   [館藏目錄]   至系統瀏覽論文 (2028-7-27以後開放)
摘要(中) 幹細胞於再生醫學以及組織工程之應用上是相當具有前瞻性的研究來源。其中,人類胚胎幹細胞以其能保有細胞原有之多功能特性而常用於生醫領域上之研究。常見的塗層培養材料,如:基質膠、層黏蛋白511、層黏蛋白521、重組玻連蛋白等等。這些塗層材料皆是以細胞分泌之細胞外間質而來。在本研究中,有三組不同的奈米片段設計,用以探索人類胚胎幹細胞於無異種材料上之生長,包含:(一)以聚(乙二醇)2-氨基乙基醚乙酸交聯劑接枝於聚(乙烯醇 - 共 - 衣康酸)水凝膠與寡玻?之間之材料 (二)於聚(乙烯醇 - 共 - 衣康酸)水凝膠接枝含不同長度之結合片段之寡?之材料 (三)可塗層於培養盤之奈米片段設計之材料,以上材料將透過鑑定分析評估其在生物醫學領域上之應用。
(一)以聚(乙二醇)2-氨基乙基醚乙酸交聯劑接枝於聚(乙烯醇 - 共 - 衣康酸)水凝膠與寡玻?間,為了使先前的研究能得以改善其表面所需寡?濃度高之問題(高於500μg/ml),本研究中利用交聯劑特性於表面提供了一長鏈結構,一則使得表面結構更加地具有彈性,便於細胞貼附時更容易找到寡?上之鍵結部位,使得表面所需之寡?含量降低。二則能模仿如同細胞外間質蛋白上之微環境供細胞更好地貼附及生長。
(二)於聚(乙烯醇 - 共 - 衣康酸)水凝膠接枝含不同長度之結合片段之寡?之設計,包含:單鏈寡?、含有結合片段之單鏈寡?、雙鏈寡?以及含有結合片段之雙鏈寡?,透過設計以上序列以探索不同結合片段於寡?上所造成之影響,並藉由人類胚胎幹細胞培養於此材料上做多能性測試以及誘導成為心肌細胞以評估其在無異種環境下生醫領域之應用特性。
(三)為了改善小分子寡?接枝於聚(乙烯醇 - 共 - 衣康酸)水凝膠時所需之高濃度用量(高於500μg/ml),希冀將疏水特性之寡?鏈連接於寡玻?之上以此模仿細胞外間質能塗層於培養盤之表面的特性,提供一種更為方便製作培養盤之方法。
以上奈米片段分子之設計,提供了三種於使用小分子寡玻?製作培養盤時之改良方法,以能使得小分子寡?能更好地運用於幹細胞培養以及再生醫學之臨床應用,並透過X射線光電子能譜、細胞型態之觀察以及免疫蛋白染色等,完整的評估其特性。
摘要(英) Stem cells are an attractive prospect for regenerative medicine and tissue engineering. The dishes coated with Matrigel, Synthemax II, Laminin 511, Laminin 521, and Cellstart are typically used for the cell culture substrates for human embryonic stem cells (hESCs). These coating materials were based on extracellular matrix (ECM), which were secreted from cells. Our previous study showed that hESCs can be cultured on polyvinylalcohol-co-itaconic acid (PVA-IA) hydrogels conjugated with higher concentration of oligovitronectin (>500 μg/mL) as hESC culture substrates. In order to solve the reason of the higher concentration usage of oligovitronectin conjugated on PVA-IA hydrogels, we designed another PVA-IA hydrogels conjugated with oligovitronectin for hESCs culture substrates using crosslinker of PEG-AEAC (Poly(ethylene glycol)-2-aminoethyl ether acetic acid) and we evaluated hESC attachment and pluripotency cultured on these hydrogels. The PVA-IA hydrogels were prepared by coating aqueous PVA-IA solution on the tissue culture polystyrene (TCPS) dishes and were crosslinked with glutaraldehyde to control hydrogel stiffness. Twice activation was performed to conjugate oligovitronectin on PVA-IA hydrogels via PEG-AEAC crosslinker by the peptide bonding reaction between carboxylic group and amino group. We investigated the effect of long crosslinking agent of PEG-AEAC on the culture of hESCs on PVA-IA hydrogels grafted with oligovitronectin via PEG-AEAC. We have also tried same reaction but with different length of PEG-AEAC to evaluate the length effect on the culture of hESCs in the dishes. After the long term culture of hESCs on the PVA-IA hydrogels (more than 10 passages), we evaluated pluripotency and differentiation ability of hESCs. Moreover, we design several oligopeptide sequence with different length of joint segment to investigate which oligopeptide-grafted hydrogel molecular design with optimal length of joint segments supported the proliferation of hESCs while hESCs maintained their pluripotency and differentiation ability into cardiomyocytes under xeno-free cell culture conditions. In addition, we designed the small molecules of oligopeptides for hESC culture, which could be coated directly to the TCPS dishes in order to decrease the concentration of these small molecules of oligopeptides used and also to provide a much more convenient way to prepare hESC culture dishes.
關鍵字(中) ★ 人類胚胎幹細胞
★ 奈米片段
★ 細胞培養
★ 無異種
★ 分子設計
★ 生醫材料
關鍵字(英) ★ human embryonic stem cells
★ molecular design
★ nanosegments
★ xeno-free
★ cell culture
★ biomaterials
論文目次 Index of Content
Abstract I
Index of Content IV
Index of Figure VII
Index of Table XII
Chapter 1 Introduction 1
1.1 Stem cells 1
1.1.1 Human Embryonic Stem Cells (hESCs) 1
1.1.2 Human Induced Pluripotent Stem Cells (hiPSCs) 3
1.2 Expansion of Human Pluripotent Stem Cells under Xeno-Free Conditions 5
1.2.1 hPSCs Cultured on Surface Coated with ECM and ECM-mimicking Peptides in 2-D Culture 6
1.2.2 hPSCs Cultured on Oligopeptides-Immobilized Surface in 2-D Culture 11
1.2.3 hPSCs Cultured on Synthetic Polymer Surface in 2-D 15
1.3 Microenvironment Effects on hPSCs 16
1.3.1 The Effects of Biomaterials Elasticity for Differentiation Fates of hPSCs 17
1.3.2 The Effects of Biomaterial Hydrophilicity on hPSC Adhesion and Function 19
1.3.3 Cell-Cell Interaction 20
1.4 Characterization of hPSCs 22
1.4.1 Colony formation 23
1.4.2 Alkali Phosphatase Activity (ALP) 24
1.4.3 Pluripotent Gene Expression 24
1.4.4 Pluripotent Protein Expression 24
1.4.5 Differentiation Ability 25
1.4.6 Immunofluorescence 27
1.5 Differentiation of hPSCs into Cardiomyocytes 28
1.5.1 Efficient Methods for Differentiating hPSCs into Cardiomyocytes 29
1.5.2 Effect of Cell Culture Biomaterials on hPSC Differentiation into Cardiomyocytes 32
1.6 The goal of this study 33
Chapter 2 Materials and Method 35
2.1 Materials 35
2.1.1 Cell Line 35
2.1.2 Commercial Culture Dishes 35
2.1.3 Commercial Coated Substrates 35
2.1.4 Medium and Others 35
2.1.5 Chemical Materials 36
2.1.6 Immunostaining 47
2.2 Cell culture 48
2.2.1 Preparation for PVA Hydrogel Dishes Conjugated with PEG Crosslinker and Oligo-peptides 48
2.2.2 Preparation for PVA-IA Hydrogels Grafted with Oligo-peptides Having Different Length of Joint Segment 50
2.2.3 Preparation for Oligo-peptides Nanosegments Coating Dishes 52
2.2.4 Culture Method of hESCs 52
2.2.5 Passage Method of hESCs 53
2.2.6 Cryopreservation of hESCs 53
2.2.7 hESCs Thawing 54
2.3 Characterization of Dish Surface 55
2.3.1 Attenuated Total Reflectance-Fourier Transform Infrared (ATR-FTIR) Spectroscopy 55
2.3.2 X-ray Photoelectron Spectroscopy (XPS) 55
2.3.3 Dynamic Light Scattering (DLS) 56
2.4 Characterization of hESCs 56
2.4.1 Expansion Fold and Doubling Time of hPSCs 56
2.4.2 Differentiation Ratio of hPSCs 57
2.4.3 Immunostaining of Cells 57
2.4.4 Embryoid Body Formation 59
2.4.5 Teratoma Formation 60
2.5 Cardiomyocyte Differentiation 61
2.5.1 Cardiomyocyte Differentiation Method 61
2.5.2 Flow Cytometry Analysis 62
Chapter 3 Results and Discussion 63
3.1 PVA-IA Hydrogel Dishes Conjugated with PEG Crosslinker and Oligopeptides 63
3.1.1 Characterization of PVA-IA Hydrogels Conjugated with PEG Crosslinker and Oligopeptides 63
3.1.2 Cultivation of hESCs on PVA-IA Hydrogels Conjugated with PEG Crosslinker and Oligo-peptides 71
3.2 PVA-IA Hydrogels Grafted with Oligo-peptides Having Different Length of Joint Segment 78
3.2.1 Characterization of PVA-IA Hydrogels Conjugated with PEG Crosslinker and Oligopeptides 78
3.2.2 Cultivation of hESCs on PVA-IA Hydrogels Grafted with Oligo-peptides Having Different Length of Joint Segment 84
3.3 Oligopeptides Nanosegments Coating Dishes 107
3.3.1 Characterization of Oligopeptides Nanosegments Coating Dishes 107
3.3.2 Cultivation of hESCs on Oligopeptides Nanosegments Coating Dishes 122
Chapter 4 Conclusion 128
Supplementary Data 131
Reference 141
參考文獻 [1] A. Higuchi et al., "Stem cell therapies for myocardial infarction in clinical trials: bioengineering and biomaterial aspects," Laboratory Investigation, 2017, vol. 97, no. 10, p. 1167.
[2] J. A. Thomson et al., "Embryonic stem cell lines derived from human blastocysts," science, 1998, vol. 282, no. 5391, pp. 1145-1147.
[3] D. A. Robinton and G. Q. Daley, "The promise of induced pluripotent stem cells in research and therapy," Nature, 2012, vol. 481, no. 7381, p. 295.
[4] A.-J. F. Carr, M. J. Smart, C. M. Ramsden, M. B. Powner, L. da Cruz, and P. J. Coffey, "Development of human embryonic stem cell therapies for age-related macular degeneration," Trends in neurosciences, 2013, vol. 36, no. 7, pp. 385-395.
[5] S. Kriks et al., "Dopamine neurons derived from human ES cells efficiently engraft in animal models of Parkinson’s disease," Nature, 2011, vol. 480, no. 7378, p. 547.
[6] M. Idelson et al., "Directed differentiation of human embryonic stem cells into functional retinal pigment epithelium cells," Cell stem cell, 2009, vol. 5, no. 4, pp. 396-408.
[7] S. D. Schwartz et al., "Human embryonic stem cell-derived retinal pigment epithelium in patients with age-related macular degeneration and Stargardt′s macular dystrophy: follow-up of two open-label phase 1/2 studies," The Lancet, 2015, vol. 385, no. 9967, pp. 509-516.
[8] J. J. Chong et al., "Human embryonic-stem-cell-derived cardiomyocytes regenerate non-human primate hearts," Nature, 2014, vol. 510, no. 7504, p. 273.
[9] S. Bhattacharya et al., "High efficiency differentiation of human pluripotent stem cells to cardiomyocytes and characterization by flow cytometry," Journal of visualized experiments: JoVE, 2014, no. 91.
[10] F. W. Pagliuca et al., "Generation of functional human pancreatic β cells in vitro," Cell, 2014, vol. 159, no. 2, pp. 428-439.
[11] A. Rezania et al., "Reversal of diabetes with insulin-producing cells derived in vitro from human pluripotent stem cells," Nature biotechnology, 2014, vol. 32, no. 11, p. 1121.
[12] A. Higuchi et al., "Stem Cell Therapies for Reversing Vision Loss," Trends in biotechnology, 2017.
[13] N. Findikli, N. Candan, and S. Kahraman, "Human embryonic stem cell culture: current limitations and novel strategies," Reproductive biomedicine online, 2006, vol. 13, no. 4, pp. 581-590.
[14] S. Yamanaka, "Induced pluripotent stem cells: past, present, and future," Cell stem cell, 2012, vol. 10, no. 6, pp. 678-684.
[15] K. Takahashi and S. Yamanaka, "Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors," cell, 2006, vol. 126, no. 4, pp. 663-676.
[16] V. K. Singh, M. Kalsan, N. Kumar, A. Saini, and R. Chandra, "Induced pluripotent stem cells: applications in regenerative medicine, disease modeling, and drug discovery," Frontiers in cell and developmental biology, 2015, vol. 3, p. 2.
[17] A. D. Celiz et al., "Materials for stem cell factories of the future," Nature materials, 2014, vol. 13, no. 6, p. 570.
[18] A. M. Ross, H. Nandivada, A. L. Ryan, and J. Lahann, "Synthetic substrates for long-term stem cell culture," Polymer, 2012, vol. 53, no. 13, pp. 2533-2539.
[19] A. Higuchi et al., "Design of polymeric materials for culturing human pluripotent stem cells: Progress toward feeder-free and xeno-free culturing," Progress in Polymer Science, 2014, vol. 39, no. 7, pp. 1348-1374.
[20] A. Higuchi, Q.-D. Ling, S.-T. Hsu, and A. Umezawa, "Biomimetic cell culture proteins as extracellular matrices for stem cell differentiation," Chemical reviews, 2012, vol. 112, no. 8, pp. 4507-4540.
[21] M. T. Nguyen et al., "Differentiation of human embryonic stem cells to endothelial progenitor cells on laminins in defined and xeno-free systems," Stem cell reports, 2016, vol. 7, no. 4, pp. 802-816.
[22] S. Rodin et al., "Clonal culturing of human embryonic stem cells on laminin-521/E-cadherin matrix in defined and xeno-free environment," Nature communications, 2014, vol. 5, p. 3195.
[23] K. Tano, S. Yasuda, T. Kuroda, H. Saito, A. Umezawa, and Y. Sato, "A novel in vitro method for detecting undifferentiated human pluripotent stem cells as impurities in cell therapy products using a highly efficient culture system," PloS one, 2014, vol. 9, no. 10, p. e110496.
[24] G. Chen et al., "Chemically defined conditions for human iPSC derivation and culture," Nature methods, 2011, vol. 8, no. 5, p. 424.
[25] Y.-M. Chen et al., "Xeno-free culture of human pluripotent stem cells on oligopeptide-grafted hydrogels with various molecular designs," Scientific reports, 2017, vol. 7, p. 45146.
[26] S. R. Braam et al., "Recombinant Vitronectin Is a Functionally Defined Substrate That Supports Human Embryonic Stem Cell Self?Renewal via αVβ5 Integrin," Stem cells, 2008, vol. 26, no. 9, pp. 2257-2265.
[27] M. Cooke et al., "Neural differentiation regulated by biomimetic surfaces presenting motifs of extracellular matrix proteins," Journal of Biomedical Materials Research Part A, 2010, vol. 93, no. 3, pp. 824-832.
[28] K. M. Hennessy 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, vol. 30, no. 10, pp. 1898-1909.
[29] R. S. Bhatnagar, J. J. Qian, and C. A. Gough, "The Role in Cell Binding of a β1-bend within the Triple Helical Region in Collagen αl (I) Chain: Structural and Biological Evidence for Conformational Tautomerism on Fiber Surface," Journal of Biomolecular Structure and Dynamics, 1997, vol. 14, no. 5, pp. 547-560.
[30] F. Gelain, D. Bottai, A. Vescovi, and S. Zhang, "Designer self-assembling peptide nanofiber scaffolds for adult mouse neural stem cell 3-dimensional cultures," PloS one, 2006, vol. 1, no. 1, p. e119.
[31] C. G. Knight et al., "Identification in collagen type I of an integrin α2β1-binding site containing an essential GER sequence," Journal of Biological Chemistry, 1998, vol. 273, no. 50, pp. 33287-33294.
[32] C. G. Knight, L. F. Morton, A. R. Peachey, D. S. Tuckwell, R. W. Farndale, and M. J. Barnes, "The Collagen-binding A-domains of Integrins α1β1 and α2β1recognize the same specific amino acid sequence, GFOGER, in native (Triple-helical) collagens," Journal of Biological Chemistry, 2000, vol. 275, no. 1, pp. 35-40.
[33] J. George, Y. Kuboki, and T. Miyata, "Differentiation of mesenchymal stem cells into osteoblasts on honeycomb collagen scaffolds," Biotechnology and bioengineering, 2006, vol. 95, no. 3, pp. 404-411.
[34] R. M. Salasznyk, W. A. Williams, A. Boskey, A. Batorsky, and G. E. Plopper, "Adhesion to vitronectin and collagen I promotes osteogenic differentiation of human mesenchymal stem cells," BioMed Research International, 2004 vol. 2004, no. 1, pp. 24-34.
[35] A. Higuchi et al., "Polymeric materials for ex vivo expansion of hematopoietic progenitor and stem cells," Journal of Macromolecular ScienceR, Part C: Polymer Reviews, 2009, vol. 49, no. 3, pp. 181-200.
[36] X.-S. Jiang, C. Chai, Y. Zhang, R.-X. Zhuo, H.-Q. Mao, and K. W. Leong, "Surface-immobilization of adhesion peptides on substrate for ex vivo expansion of cryopreserved umbilical cord blood CD34+ cells," Biomaterials, 2006, vol. 27, no. 13, pp. 2723-2732.
[37] R. Bhatia, A. D. Williams, and H. A. Munthe, "Contact with fibronectin enhances preservation of normal but not chronic myelogenous leukemia primitive hematopoietic progenitors," Experimental hematology, 2002, vol. 30, no. 4, pp. 324-332.
[38] Z. Melkoumian et al., "Synthetic peptide-acrylate surfaces for long-term self-renewal and cardiomyocyte differentiation of human embryonic stem cells," Nature biotechnology, 2010, vol. 28, no. 6, p. 606.
[39] M. D. Pierschbacher and E. Ruoslahti, "Cell attachment activity of fibronectin can be duplicated by small synthetic fragments of the molecule," Nature, 1984 vol. 309, no. 5963, pp. 30-33.
[40] I.-S. Park et al., "The correlation between human adipose-derived stem cells differentiation and cell adhesion mechanism," Biomaterials, 2009, vol. 30, no. 36, pp. 6835-6843.
[41] M. Nomizu et al., "Cell binding sequences in mouse laminin α1 chain," Journal of Biological Chemistry, 1998, vol. 273, no. 49, pp. 32491-32499.
[42] K. Saha, E. F. Irwin, J. Kozhukh, D. V. Schaffer, and K. E. Healy, "Biomimetic interfacial interpenetrating polymer networks control neural stem cell behavior," Journal of Biomedical Materials Research Part A, 2007, vol. 81, no. 1, pp. 240-249.
[43] S. Suzuki, A. Oldberg, E. G. Hayman, M. D. Pierschbacher, and E. Ruoslahti, "Complete amino acid sequence of human vitronectin deduced from cDNA. Similarity of cell attachment sites in vitronectin and fibronectin," The EMBO journal, 1985, vol. 4, no. 10, pp. 2519-2524.
[44] A. Oldberg, A. Franzen, D. Heinegard, M. Pierschbacher, and E. Ruoslahti, "Identification of a bone sialoprotein receptor in osteosarcoma cells," Journal of Biological Chemistry, 1988, vol. 263, no. 36, pp. 19433-19436.
[45] J. R. Klim, L. Li, P. J. Wrighton, M. S. Piekarczyk, and L. L. Kiessling, "A defined glycosaminoglycan-binding substratum for human pluripotent stem cells," Nature methods, 2010, vol. 7, no. 12, p. 989.
[46] P. Kolhar, V. R. Kotamraju, S. T. Hikita, D. O. Clegg, and E. Ruoslahti, "Synthetic surfaces for human embryonic stem cell culture," Journal of biotechnology, 2010, vol. 146, no. 3, pp. 143-146.
[47] G. Meng, S. Liu, and D. E. Rancourt, "Synergistic effect of medium, matrix, and exogenous factors on the adhesion and growth of human pluripotent stem cells under defined, xeno-free conditions," Stem cells and development, 2011, vol. 21, no. 11, pp. 2036-2048.
[48] N. Nishishita, M. Shikamura, C. Takenaka, N. Takada, N. Fusak, and S. Kawamata, "Generation of virus-free induced pluripotent stem cell clones on a synthetic matrix via a single cell subcloning in the naive state," PloS one, 2012, vol. 7, no. 6, p. e38389.
[49] D. A. Brafman, C. W. Chang, A. Fernandez, K. Willert, S. Varghese, and S. Chien, "Long-term human pluripotent stem cell self-renewal on synthetic polymer surfaces," Biomaterials, 2010, vol. 31, no. 34, pp. 9135-9144.
[50] L. G. Villa-Diaz et al., "Synthetic polymer coatings for long-term growth of human embryonic stem cells," Nature biotechnology, 2010, vol. 28, no. 6, p. 581.
[51] E. F. Irwin, R. Gupta, D. C. Dashti, and K. E. Healy, "Engineered polymer-media interfaces for the long-term self-renewal of human embryonic stem cells," Biomaterials, 2011, vol. 32, no. 29, pp. 6912-6919.
[52] A. Higuchi, Q.-D. Ling, Y. Chang, S.-T. Hsu, and A. Umezawa, "Physical cues of biomaterials guide stem cell differentiation fate," Chemical reviews, 2013, vol. 113, no. 5, pp. 3297-3328.
[53] A. Higuchi et al., "Physical cues of cell culture materials lead the direction of differentiation lineages of pluripotent stem cells," Journal of Materials Chemistry B, 2015, vol. 3, no. 41, pp. 8032-8058.
[54] A. Higuchi et al., "Long-term xeno-free culture of human pluripotent stem cells on hydrogels with optimal elasticity," Scientific reports, 2015, vol. 5, p. 18136.
[55] J. Zoldan, E. D. Karagiannis, C. Y. Lee, D. G. Anderson, R. Langer, and S. Levenberg, "The influence of scaffold elasticity on germ layer specification of human embryonic stem cells," Biomaterials, 2011, vol. 32, no. 36, pp. 9612-9621.
[56] C. Chung, E. Anderson, R. R. Pera, B. L. Pruitt, and S. C. Heilshorn, "Hydrogel crosslinking density regulates temporal contractility of human embryonic stem cell-derived cardiomyocytes in 3D cultures," Soft matter, 2012, vol. 8, no. 39, pp. 10141-10148.
[57] A. Arshi et al., "Rigid microenvironments promote cardiac differentiation of mouse and human embryonic stem cells," Science and technology of advanced materials, 2013, vol. 14, no. 2, p. 025003.
[58] L. B. Hazeltine, M. G. Badur, X. Lian, A. Das, W. Han, and S. P. Palecek, "Temporal impact of substrate mechanics on differentiation of human embryonic stem cells to cardiomyocytes," Acta biomaterialia, 2014, vol. 10, no. 2, pp. 604-612.
[59] Y. Mei et al., "Combinatorial development of biomaterials for clonal growth of human pluripotent stem cells," Nature materials, 2010, vol. 9, no. 9, p. 768.
[60] Y. Tamada and Y. Ikada, "Effect of preadsorbed proteins on cell adhesion to polymer surfaces," Journal of colloid and interface science, 1993, vol. 155, no. 2, pp. 334-339.
[61] D. Vi?nji? and V. Crljen-Manestar, "The Cell: a molecular approach, (Geoffrey M Cooper and Robert E Hausman) chapter 13: Cell Signaling," 2004.
[62] B. M. Gumbiner, "Regulation of cadherin-mediated adhesion in morphogenesis," Nature reviews Molecular cell biology, 2005, vol. 6, no. 8, p. 622.
[63] P. Das, N. B. Chandar, S. Chourey, H. Agarwalla, B. Ganguly, and A. Das, "Role of metal ion in specific recognition of pyrophosphate ion under physiological conditions and hydrolysis of the phosphoester linkage by alkaline phosphatase," Inorganic chemistry, 2013, vol. 52, no. 19, pp. 11034-11041.
[64] R. Morton, "Transferase activity of hydrolytic enzymes," Nature, 1953, vol. 172, no. 4367, p. 65.
[65] G. Ciarimboli and E. Schlatter, "Regulation of organic cation transport," Pflugers Archiv, 2005, vol. 449, no. 5, pp. 423-441.
[66] M. Bao, X. Wang, H. Yuan, X. Lou, Q. Zhao, and Y. Zhang, "HAp incorporated ultrafine polymeric fibers with shape memory effect for potential use in bone screw hole healing," Journal of Materials Chemistry B, 2016, vol. 4, no. 31, pp. 5308-5320.
[67] T. C. Schumacher et al., "Synthesis and mechanical evaluation of Sr-doped calcium-zirconium-silicate (baghdadite) and its impact on osteoblast cell proliferation and ALP activity," Biomedical Materials, 2015, vol. 10, no. 5, p. 055013.
[68] Z. Ahmad and K.-P. Huang, "Dephosphorylation of rabbit skeletal muscle glycogen synthase (phosphorylated by cyclic AMP-independent synthase kinase 1) by phosphatases," Journal of Biological Chemistry, 1981, vol. 256, no. 2, pp. 757-760.
[69] B. Brugg and A. Matus, "Phosphorylation determines the binding of microtubule-associated protein 2 (MAP2) to microtubules in living cells," The Journal of Cell Biology, 1991, vol. 114, no. 4, pp. 735-743.
[70] Z. Saiyed, S. Sharma, R. Godawat, S. Telang, and C. Ramchand, "Activity and stability of alkaline phosphatase (ALP) immobilized onto magnetic nanoparticles (Fe3O4)," Journal of biotechnology, 2007, vol. 131, no. 3, pp. 240-244.
[71] T. Miyamoto, C. Furusawa, and K. Kaneko, "Pluripotency, differentiation, and reprogramming: A gene expression dynamics model with epigenetic feedback regulation," PLoS computational biology, 2015, vol. 11, no. 8, p. e1004476.
[72] A. Higuchi, Q.-D. Ling, Y.-A. Ko, Y. Chang, and A. Umezawa, "Biomaterials for the feeder-free culture of human embryonic stem cells and induced pluripotent stem cells," Chemical reviews, 2011, vol. 111, no. 5, pp. 3021-3035.
[73] S. Rungarunlert, M. Techakumphu, M. K. Pirity, and A. Dinnyes, "Embryoid body formation from embryonic and induced pluripotent stem cells: Benefits of bioreactors," World journal of stem cells, 2009, vol. 1, no. 1, p. 11.
[74] R. V. Nelakanti, N. G. Kooreman, and J. C. Wu, "Teratoma formation: a tool for monitoring pluripotency in stem cell research," Current protocols in stem cell biology, 2015, pp. 4A. 8.1-4A. 8.17.
[75] I. D. Odell and D. Cook, "Immunofluorescence techniques," The Journal of investigative dermatology, 2013, vol. 133, no. 1, p. e4.
[76] R. C. Ladner, "Mapping the epitopes of antibodies," Biotechnology and Genetic Engineering Reviews, 2007, vol. 24, no. 1, pp. 1-30.
[77] R. Mandrell, J. Griffiss, and B. Macher, "Lipooligosaccharides (LOS) of Neisseria gonorrhoeae and Neisseria meningitidis have components that are immunochemically similar to precursors of human blood group antigens. Carbohydrate sequence specificity of the mouse monoclonal antibodies that recognize crossreacting antigens on LOS and human erythrocytes," Journal of Experimental Medicine, 1988, vol. 168, no. 1, pp. 107-126.
[78] K. R. Boheler, J. Czyz, D. Tweedie, H.-T. Yang, S. V. Anisimov, and A. M. Wobus, "Differentiation of pluripotent embryonic stem cells into cardiomyocytes," Circulation research, 2002, vol. 91, no. 3, pp. 189-201.
[79] H. Kempf, C. Kropp, R. Olmer, U. Martin, and R. Zweigerdt, "Cardiac differentiation of human pluripotent stem cells in scalable suspension culture," Nature protocols, 2015, vol. 10, no. 9, p. 1345.
[80] C. L. Mummery, J. Zhang, E. S. Ng, D. A. Elliott, A. G. Elefanty, and T. J. Kamp, "Differentiation of human embryonic stem cells and induced pluripotent stem cells to cardiomyocytes: a methods overview," Circulation research, 2012, vol. 111, no. 3, pp. 344-358.
[81] P. W. Burridge et al., "Chemically defined generation of human cardiomyocytes," Nature methods, vol. 11, no. 8, p. 855, 2014.
[82] P. Menasche et al., "Towards a clinical use of human embryonic stem cell-derived cardiac progenitors: a translational experience," European heart journal, 2014, vol. 36, no. 12, pp. 743-750.
[83] I. Y. Shadrin et al., "Cardiopatch platform enables maturation and scale-up of human pluripotent stem cell-derived engineered heart tissues," Nature communications, 2017, vol. 8, no. 1, p. 1825.
[84] A. Higuchi et al., "Polymeric design of cell culture materials that guide the differentiation of human pluripotent stem cells," Progress in Polymer Science, 2017, vol. 65, pp. 83-126.
[85] N. C. Dubois et al., "SIRPA is a specific cell-surface marker for isolating cardiomyocytes derived from human pluripotent stem cells," Nature biotechnology, 2011, vol. 29, no. 11, p. 1011.
[86] X. Lian et al., "Directed cardiomyocyte differentiation from human pluripotent stem cells by modulating Wnt/β-catenin signaling under fully defined conditions," Nature protocols, 2013, vol. 8, no. 1, p. 162.
[87] A. Mihic et al., "The effect of cyclic stretch on maturation and 3D tissue formation of human embryonic stem cell-derived cardiomyocytes," Biomaterials, 2014, vol. 35, no. 9, pp. 2798-2808.
[88] D. Zhang, I. Y. Shadrin, J. Lam, H.-Q. Xian, H. R. Snodgrass, and N. Bursac, "Tissue-engineered cardiac patch for advanced functional maturation of human ESC-derived cardiomyocytes," Biomaterials, 2013, vol. 34, no. 23, pp. 5813-5820.
[89] M. Lecina, S. Ting, A. Choo, S. Reuveny, and S. Oh, "Scalable platform for human embryonic stem cell differentiation to cardiomyocytes in suspended microcarrier cultures," Tissue Engineering Part C: Methods, 2010, vol. 16, no. 6, pp. 1609-1619.
[90] A. J. Ribeiro et al., "Contractility of single cardiomyocytes differentiated from pluripotent stem cells depends on physiological shape and substrate stiffness," Proceedings of the National Academy of Sciences, 2015, vol. 112, no. 41, pp. 12705-12710.
[91] M. R. Salick et al., "Micropattern width dependent sarcomere development in human ESC-derived cardiomyocytes," Biomaterials, 2014, vol. 35, no. 15, pp. 4454-4464.
[92] M. J. Birket et al., "Contractile defect caused by mutation in MYBPC3 revealed under conditions optimized for human PSC-cardiomyocyte function," Cell reports, 2015, vol. 13, no. 4, pp. 733-745.
[93] T.-J. Lee et al., "Incorporation of gold-coated microspheres into embryoid body of human embryonic stem cells for cardiomyogenic differentiation," Tissue Engineering Part A, 2014, vol. 21, no. 1-2, pp. 374-381.
[94] B. Jiang et al., "Generation of cardiac spheres from primate pluripotent stem cells in a small molecule-based 3D system," Biomaterials, 2015, vol. 65, pp. 103-114.
[95] K. Shapira-Schweitzer, M. Habib, L. Gepstein, and D. Seliktar, "A photopolymerizable hydrogel for 3-D culture of human embryonic stem cell-derived cardiomyocytes and rat neonatal cardiac cells," Journal of molecular and cellular cardiology, 2009, vol. 46, no. 2, pp. 213-224.
[96] R. E. Horton and D. T. Auguste, "Synergistic effects of hypoxia and extracellular matrix cues in cardiomyogenesis," Biomaterials, 2012, vol. 33, no. 27, pp. 6313-6319.
[97] X. Lian et al., "Robust cardiomyocyte differentiation from human pluripotent stem cells via temporal modulation of canonical Wnt signaling," Proceedings of the National Academy of Sciences, 2012, vol. 109, no. 27, pp. E1848-E1857.
[98] S. Ting, A. Chen, S. Reuveny, and S. Oh, "An intermittent rocking platform for integrated expansion and differentiation of human pluripotent stem cells to cardiomyocytes in suspended microcarrier cultures," Stem cell research, 2014, vol. 13, no. 2, pp. 202-213.
[99] R. Jha et al., "Molecular beacon-based detection and isolation of working-type cardiomyocytes derived from human pluripotent stem cells," Biomaterials, 2015, vol. 50, pp. 176-185.
[100] N. Huebsch et al., "Automated video-based analysis of contractility and calcium flux in human-induced pluripotent stem cell-derived cardiomyocytes cultured over different spatial scales," Tissue Engineering Part C: Methods, 2015, vol. 21, no. 5, pp. 467-479.
[101] Y. W. Chun et al., "Combinatorial polymer matrices enhance in vitro maturation of human induced pluripotent stem cell-derived cardiomyocytes," Biomaterials, 2015, vol. 67, pp. 52-64.
[102] L. Ikonen, E. Kerkela, G. Metselaar, M. C. Stuart, M. R. De Jong, and K. Aalto-Setala, "2D and 3D self-assembling nanofiber hydrogels for cardiomyocyte culture," BioMed research international, 2013, vol. 2013.
[103] J. Dahlmann et al., "The use of agarose microwells for scalable embryoid body formation and cardiac differentiation of human and murine pluripotent stem cells," Biomaterials, 2013, vol. 34, no. 10, pp. 2463-2471.
[104] D. Jing, A. Parikh, and E. S. Tzanakakis, "Cardiac cell generation from encapsulated embryonic stem cells in static and scalable culture systems," Cell transplantation, 2010, vol. 19, no. 11, pp. 1397-1412.
[105] A. Higuchi and T. Iijima, "DSC investigation of the states of water in poly (vinyl alcohol) membranes," Polymer, 1985, vol. 26, no. 8, pp. 1207-1211.
[106] A. Higuchi and T. Iijima, "DSC investigation of the states of water in poly (vinyl alcohol-co-itaconic acid) membranes," Polymer, 1985, vol. 26, no. 12, pp. 1833-1837.
[107] L. D. Wilsbacher and S. R. Coughlin, "Analysis of cardiomyocyte development using immunofluorescence in embryonic mouse heart," Journal of visualized experiments: JoVE, 2015, no. 97.
[108] ThermoScientific,"NHS and Sulfo-NHS instructions"Home Page: https://assets.thermofisher.com/TFS-Assets/LSG/manuals/MAN0011309_NHS_SulfoNHS_UG.pdf, 2018.
指導教授 ?口亞紺(Akon Higuchi) 審核日期 2018-7-30
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