博碩士論文 973204008 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:19 、訪客IP:3.14.6.194
姓名 黃曉茜(Shiau-Chian Huang)  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 微環境對羊水間葉幹細胞多功能性基因表現及分化之影響
(Effect of Micro-environment on Pluripotent Gene Expression and Differentiation of Mesenchymal Stem Cell from Second-trimester Amniotic Fluid)
相關論文
★ 於不同彈性係數的生醫材料上體外培植造血幹細胞★ 藉由調整水凝膠之表面電荷及軟硬度並嫁接玻連蛋白用以培養人類多功能幹細胞
★ 可見光對羊水間葉幹細胞成骨分化之影響★ 可見光調控神經細胞之基因表現及突觸生長
★ 膜純化法及免疫抗體磁珠法用於分離及體外增殖血液幹細胞之研究★ 人類表皮成長因子的結構穩定性及生物活性測定
★ 奈米片段與細胞外基質之改質膜用於臍帶血中造血幹細胞之純化與培養★ 小鼠脂肪幹細胞之膜純化法及細胞外間質對人類脂肪幹細胞影響之研究
★ 利用具有奈米片段與細胞外間質蛋白質的表面改殖材質進行臍帶血造血幹細胞體外培養★ 在不同培養條件下針對大腸癌細胞及組織中癌細胞進行純化、剔除及鑑定之研究
★ 羊水間葉幹細胞培養於細胞外間質改質表面其分化能力及多能性之研究★ 人類脂肪幹細胞的膜純化法與分化能力研究
★ 具有抗藥性之大腸癌細胞株能提高癌胚抗原的表現,但並非是癌症起始細胞★ 羊水間葉幹細胞培養於接枝細胞外間質寡肽與環狀肽具有最佳表面硬度的生醫材料,其增殖能力及多能性之研究
★ 人類體細胞從組成誘導型多能性幹細胞培養在無飼養層上★ 使用不同孔洞大小之耐倫薄膜從脂肪組織中分離及純化人類脂肪幹細胞之研究
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) 間葉幹細胞在臨床應用上為最方便取得且可自我細胞移植的幹細胞來源之ㄧ,但間葉幹細胞其分化能力不如胚胎幹細胞。近年來有許多研究發現從懷孕婦女的羊水中純化出間葉幹細胞,而純化出的間葉幹細胞為多功能幹細胞並具有像胚胎幹細胞能力可分化三種胚層的細胞。由於微環境 (例如:培養液和培養皿接枝不同官能基或細胞外間質蛋白質) 在間葉幹細胞的基因表現與分化能力有極大的影響,我們將懷孕15至25周的羊水培養於不同的表面材質中,例如電漿改質接上不同的官能基、塗佈或者化學鍵結固定細胞外間質蛋白質於培養皿進行純化與培養繼代,測試多功能性基因表現量與分化能力。我們發現將羊水培養於特定材質的培養皿以純化間葉幹細胞,如纖維蛋白質固定化於改質PS培養皿可得較高的多功能性基因表現,即使經由繼代依舊可維持其多功能性表現;另外,從羊水中純化出的間葉幹細胞除了可分化屬中胚層的脂肪細胞和成骨細胞以外,亦可分化屬外胚層的神經細胞,故從羊水純化之間葉幹細胞為一種多功能幹細胞,並受培養微環境而影響其多功能性基因表現及分化能力。
摘要(英) The cells derived from second-trimester amniocentesis contain pluripotent stem cells capable of differentiating into multiple lineages, including representatives of all three embryonic germ layers. Therefore, amniotic fluid may be a suitable alternative source of damaged cells in our body. Because micro-environment of stem cells (i.e., surface chemistry and topography of culture materials) plays an important role on gene expression and differentiation of stem cells, we isolated stem cells from second-trimester amniotic fluid, and cultured amniotic-derived stem cells on the surface of culture dishes immobilized several ECM proteins by coating or chemical grafting method to generate the nano-segments on the surface (e.g., gelatin, collagen, fibronectin, laminin, vitronectin, Matrigel, and synthetic polymer nano-segments having NH2 group). The culture dishes having those nano-segments were prepared by plasma-polymerization method of glycidylmethacrylate on polystyrene dishes followed by the reaction between epoxy group and ammonia to introduce NH2 groups. Immobilization of proteins on the culture dishes having NH2 groups were performed by using succinimide chemistry. The effect of interaction between amniotic-derived stem cells and nano-segments on the expression and maintenance of pluripotent genes (Oct-4, Sox-2 and Rex-1) was investigated from qRT-PCR and RT-PCR analysis. The amniotic-derived stem cells cultured on the surface of culture dishes having specific nano-segments (e.g., fibronectin) by using chemical grafting method kept pluripotent gene expression of stem cells for a long time. The differentiation ability of amniotic-derived stem cells to osteogenic, adipogenic, and neural cell lineages was also investigated. Differentiation ability to each cell lineages was also dependent on the surface chemistry of culture dishes having specific nano-segments. In conclusion, gene expression of pluripotency and specific differentiation are found to depend on the surface chemistry of culture materials having specific nano-segments.
關鍵字(中) ★ 細胞外間質
★ 多功能基因表現
★ 間葉幹細胞
關鍵字(英) ★ MSC
★ ECM
★ Pluripotent gene expression
論文目次 CHAPTER ONE INTRODUCTION ..................................................................................... 1
1-1 Stem Cell ...................................................................................................................... 1
1-1.1 Potency of stem cells................................................................................................. 1
1-1.1.1 Totipotency ........................................................................................................ 1
1-1.1.2 Pluripotency ...................................................................................................... 1
1-1.1.3 Multipotency ..................................................................................................... 2
1-1.2 Sources of stem cells ................................................................................................. 2
1-1.2.1 Embryonic stem cells (ESCs) ........................................................................... 2
1-1.2.2 Hematopoietic stem cells (HSCs) ..................................................................... 3
1-1.2.3 Mesenchymal stem cells (MSCs) ...................................................................... 4
1-2 Amniotic Fluid Cells ................................................................................................... 8
1-2.1 Amniotic fluid cell type ............................................................................................ 8
1-2.1.1 F-type colonies .................................................................................................. 9
1-2.1.2 AF-type colonies ............................................................................................... 9
1-2.1.3 E-type colonies ................................................................................................ 10
1-2.2 Isolation of amniotic fluid stem cell ....................................................................... 10
1-2.3 Characterization of amniotic fluid stem cell ........................................................... 10
1-2.4 Pluripotency of amniotic fluid stem cells ............................................................... 12
1-3 Effect of micro-environment of mesenchymal stem cells ...................................... 15
1-3.1 Culture medium effect of mesenchymal stem cell .................................................. 16
1-3.2 Effect of culture material for differentiation of mesenchymal stem cell ................ 17
1-3.2.1 Introduction of natural polymeric materials.................................................... 17
1-3.2.2 Conventional synthetic and natural polymeric materials ................................ 21
1-3.2.3 Materials manufactured with nanotechnology and having nanosegments...... 22
1-3.2.4 Application of mesnchymal stem cells ........................................................... 22
1-4 Analysis ...................................................................................................................... 24
1-4.1 X-ray photoelectron spectroscopy .......................................................................... 24
1-4.2 Flow cytometry analysis ......................................................................................... 26
1-4.3 Polymerase chain reaction (PCR) ........................................................................... 26
1-4.3.1 Procedure of PCR............................................................................................ 26
1-4.3.2 Reverse transcription polymerase chain reaction (RT-PCR) and Quantitative real
time polymerase chain reaction (qRT-PCR) ................................................................... 28
1-4.3.3 Procedure of RT-PCR ..................................................................................... 29
CHAPTER TWO MATERIALS AND METHODS ............................................................ 30
2-1 Materials .................................................................................................................... 30
2-2 Methods ...................................................................................................................... 34
2-2.1 Preparation of Cell isolation and culture................................................................. 34
2-2.2 Preparation of culture materials .............................................................................. 35
2-2.3 Analysis ................................................................................................................... 39
3-1 Analysis of materials ................................................................................................. 45
3-2 Morphology of amniotic fluid stem cells cultured on different culture dishes .... 47
3-3 Purification of stem cell from amniotic fluid .......................................................... 54
3-4 Effect of pluripotency gene expression on materials of culture dishes ................ 58
3-5 Effect of differentiation ability of AFCs on materials of culture dishes ............... 63
3-6 Summary .................................................................................................................... 89
CHAPTER FOUR CONCLUSION .................................................................................... 90
CHAPTER FIVE REFERENCES ...................................................................................... 92
CHAPTER SIX Appendix .................................................................................................. 103
參考文獻 1. Fernando Ulloa-Montoya, Catherine M. Verfaillie, and Wei-Shou Hu.” Culture Systems for Pluripotent Stem Cells”, Journal of Bioscience and Bioengineering, Vol. 100, No. 1, 12–27, (2005)
2. C. Verfaillie.” Pluripotent stem cells”, Transfusion Clinique et Biologique, (2009)
3. On line resources : Cell signal
http://www.cellsignal.com
4. On line resources︰Wikipimedia,
http://en.wikipedia.org
5. Hans R. Scholer ,"The Potential of Stem Cells: An Inventory". in Nikolaus Knoepffler, Dagmar Schipanski, and Stefan Lorenz Sorgner. Humanbiotechnology as Social Challenge. Ashgate Publishing, Ltd. 28, (2007).
6. Martin MJ, Muotri A, Gage F, Varki A, "Human embryonic stem cells express an immunogenic nonhuman sialic acid". Nat. Med. Vol. 11: 228–32, (2005).
7. Klimanskaya I, Chung Y, Meisner L, Johnson J, West MD, Lanza R, "Human embryonic stem cells derived without feeder cells". Lancet Vol. 365: 1636–41 (2005).
8. J.M. Cabrita, B.S. Ferreira, C.L. da Silva, R. Goncalves, G. Almeida-Porada and J.M.S. Cabral, “Hematopoietic stem cells: from the bone to the bioreactor”, TRENDS in Biotechnology, Vol. 21, 223-240, (2003).
9. T. Reya, S.J. Morrison, M.F. Clarke and I.L. Weissman, “Stem cells, cancer, and cancer stem cells”, Nature, Vol. 414, 105-111, (2001).
10. L Gucciardo, R Lories, N Ochsenbein-Ko-lble, E Done’, A Zwijsen, J Deprest , “Fetal mesenchymal stem cells: isolation, properties and potential use in perinatology and regenerative medicine”, Best Practice & Research Clinical Obstetrics and Gynecology,(2008).
11. Dario Fauza, “Amniotic fluid and placental stem cells”, Best Practice & Research Clinical Obstetrics and Gynecology, Vol. 18, No. 6, pp. 877–891, (2004)
12. M. Hengstschlager, “stem cell in amniotic fluid”, J Reproduktionsmed Endokrinol, Vol. 2, 233–8,(2005)
13. Yash M. Kolambkar , Alexandra Peister, Shay Soker , Anthony Atala , Robert E. Guldberg, “Chondrogenic differentiation of amniotic fluid-derived stem cells”, J Mol Hist, Vol. 38, 405–413, (2007)
14. Paolo De Coppi, Georg Bartsch, Jr, M Minhaj Siddiqui, Tao Xu, Cesar C Santos, Laura Perin, Gustavo Mostoslavsky, Ange’line C Serre, Evan Y Snyder, James J Yoo, Mark E Furth, Shay Soker &Anthony Atala, “Isolation of amniotic stem cell lines with potential for therapy”, Nature biotechnology, Vol. 25, No. 1,(2007).
15. Yu-Bao Zheng, Zhi-Liang Gao, Chan Xie, Hai-Peng Zhu, Liang Peng, Jun-Hong Chen, Yu Tian Chong, “Characterization and hepatogenic differentiation of mesenchymal stem cells from human amniotic fluid and human bone marrow: A comparative study”, Cell Biology International, In press, 1-10,(2008)
16. Ming-Song Tsai, Shiaw-Min Hwang, Yieh-Loong Tsai, Fu-Chou Cheng, Jia-Ling Lee, and Yu-Jen Chang, “Clonal Amniotic Fluid-Derived Stem Cells Express Characteristics of Both Mesenchymal and Neural Stem Cells”, Biology of Reproduction, Vol. 74, 545–551, (2006)
17. I. Virtanen, H. Von koskull, V-P. Lehto, T. Vartio, and P. Aula, “Cultured Human Amniotic Fluid Cells Characterized with Antibodies against Intermediate Filaments in Indirect Immunofluorescence Microscopy”, J. Clin. Invest., Vol. 68, 1348-1355,(1981)
18. Ming-Song Tsai, Shiaw-Min Hwang, Kuang-Den Chen, Yun-Shien Lee, Li-Wen Hsu, Yu-Jen Chang, Chao-Nin Wang, Hsiu-Huei Peng, Yao-Lung Chang, An-Shine Chao, Shuenn-Dyh Chang, Kuan-Der Lee, Tzu-Hao Wang, Hsin-Shih Wang, Yung-Kuei Soong, “Functional Network Analysis of the Transcriptomes of Mesenchymal Stem Cells Derived from Amniotic Fluid, Amniotic Membrane, Cord Blood, and Bone Marrow” Stem Cells, Vol. 25, 2511–2523,(2007).
19. J. Kim, Y. Lee, H. Kim, K. J. Hwang, H. C. Kwon, S. K. Kim, D. J. Cho, S. G. Kang and J. You, “Human amniotic fluid-derived stem cells have characteristics of multipotent stem cells”, Cell Prolif., Vol. 40, 75–90,(2007).
20. A Poloni1, V Rosini, E Mondini, G Maurizi, S Mancini, G Discepoli, S Biasio, G Battaglini, E Berardinelli, F Serrani and P Leoni, “Characterization and expansion of mesenchymal progenitor cells from first-trimester chorionic villi of human placenta”, Cytotherapy, Vol. 10, No. 7, 690-697,(2008).
21. Venkata Lokesh Battula, Petra M. Bareiss, Sabrina Treml, Sabine Conrad, Ingrid Albert, Sigrid Hojak, Harald Abele, Bernhard Schewe, Lothar Just, Thomas Skutella , “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, Vol. 75,279–291,(2007)
22. Kristine M. Safford, Kevin C. Hicok, Shawn D. Safford, Yuan-Di C. Halvorsen, William O. Wilkison, Jeffrey M. Gimble, Henry E. Rice, “Neurogenic differentiation of murine and human adipose-derived stromal cells”, Biochemical and Biophysical Research Communications, Vol. 294, 371-379, (2002)
23. S. P. Hector, R. Cavina, A. Sala, F. Latteri, E. Campagnoli, V. Ginanni, E. Morenghi, L. Rimassa, G. Biancofiore, A. Santoro, “Feasibility of combination of cisplatin (CDDP) and gemcitabine (GEM) in non-small cell lung cancer and other solid tumors: analysis of dose-intensity (DI) and compliance of four different schedules”, European Journal of Cancer, Vol. 37, Supplement 6, S64-S65, (2001)
24. Francesco Dell’Accio, Cosimo De Bari, Frank P. Luyten, “Microenvironment and phenotypic stability specify tissue formation by human articular cartilage-derived cells in vivo”, Experimental Cell Research, Vol, 287, 16-27 , (2003)
25. Gosden CM, “Amniotic fluid cell types and culture”, Br Med Bull, Vol. 39, 348–54, (1983).
26. Jiang, Y., Jahagirdar, B. N., Reinhardt, R. L., Schwartz, R. E., Keene, C. D., Ortiz-Gonzalez, X. R., Reyes, M., Lenvik, T., Lund, T., Blackstad, M., and other 6 authors: Pluripotency of mesenchymal stem cells derived from adult marrow. Nature, 418, 41–49, (2002).
27. Reyes, M., Lund, T., Lenvik, T., Aguiar, D., Koodie, L., and Verfaillie, C. M.: Purification and ex vivo expansion of postnatal human marrow mesodermal progenitor cells. Blood, 98, 2615–2625, (2001).
28. Prusa AR, Hengstschlager M, “Amniotic fluid cells and human stem cell research: a new connection”, Med Sci Monit, Vol. 8, 253–257, (2002).
29. Milunsky A, “Amniotic fluid cell culture.”, Milunsky A (ed). Genetic disorder and the fetus. Plenum Press, New York, (1979).
30. Hoehn H, Salk D, “Morphological and biochemical heterogeneity of amniotic fluid cells in culture.”, Methods Cell Biol, Vol. 26: 11–34, (1982).
31. Laura Perrone, Mario Giuffre, Caterina D'Alfonso, Maria T. Carbone, Giuseppe Presta, Antonino Di Toro, Rosario Di Toro, “Postnatal weight change is influenced by mother-newborn pair leptin levels”, Nutrition Research, Vol. 20, 1531-1536, (2000)
32. R. Salingcarnboriboon, H. Yoshitake, K. Tsuji, M. Obinata, T. Amagasa, A. Nifuji, M. Noda, “Establishment of tendon-derived cell lines exhibiting pluripotent mesenchymal stem cell-like property”, Experimental Cell Research, Vol. 287, 289-300, (2003)
33. Maria Teresa Valenti , Luca Dalle Carbonare, Luca Donatelli, Francesco Bertoldo, Mirko Zanatta, Vincenzo Lo Cascio, “Gene expression analysis in osteoblastic differentiation from peripheral blood MSC”, Bone, 43, 1084–1092, (2008)
34. Reza Izadpanah, Cynthia Trygg, Bindiya Patel, Christopher Kriedt, Jason Dufour, Jeffery M. Gimble, and Bruce A. Bunnell, “Biologic Properties of Mesenchymal Stem Cells Derived From Bone Marrow and Adipose Tissue”, Journal of Cellular Biochemistry, Vol. 99, 1285–1297, (2006)
35. Yen BL, Huang HI, Chien CC, Jui HY, Ko BS, Yao M, Shun CT, Yen ML, Lee MC, Chen YC., “Isolation of multipotent cells from human term placenta”, Stem Cells, Vol. 23(1), 3-9 , (2005)
36. Kazutoshi Takahashi and Shinya Yamanaka, “Induction of Pluripotent Stem Cells from Mouse Embryonic and Adult Fibroblast Cultures by Defined Factors”, Cell, Vol. 126, 1–14, (2006)
37. Ming-Song Tsai, Jia-Ling Lee, Yu-Jen Chang and Shiaw-Min Hwang, “Isolation of human multipotent mesenchymal stem cells from second-trimester amniotic fluid using a novel two-stage culture protocol”, Human Reproduction Vol.19, No.6, 1450-1456, (2004)
38. Fernanda Zambotti, Karl Blau, Graham S. King, Stuart Campbell, M. Sandler, “Monoamine metabolites and related compounds in human amniotic fluid: Assay by gas chromatography and gas chromatography-mass spectrometry”, Clinica Chimica Acta, Vol. 61, 247-256, (1975)
39. Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, Mosca JD, Moorman MA, Simonetti DW, Craig S, Marshak DR. “Multilineage potential of adult human mesenchymal stem cells.” Science, Vol. 284, 143–7, (1999).
40. Shamblott MJ, Axelman J, Littlefield JW, Blumenthal PD, Huggins GR, Cui Y, Cheng L, Gearhart JD. “Human embryonic germ cell derivatives express a broad range of developmentally distinct markers and proliferate extensively in vitro.”, Proc Natl Acad Sci USA, Vol. 98, 113–8 , (2001).
41. Baksh, D., Song, L., and Tuan, R. S.: Adult mesenchymal stem cells: characterization, differentiation, and application in cell and gene therapy. J. Cell. Mol. Med., 8, 301–316, (2004).
42. Andrea-Romana Prusa, Erika Marton, Margit Rosner, Gerhard Bernaschek and Markus HengstschlaEger, “Oct-4-expressing cells in human amniotic fluid-a new source for stem cell research”, Human Reproduction Vol.18, No.7, 1489-1493, (2003)
43. Zuk PA, Zhu M, Ashjian P, DeUgarte DA, Huang JI, Mizuno H, Alfonso ZC, Fraser JK, Benhaim P, Hedrick MH. “Human adipose tissue is a source of multipotent stem cells.” Mol Biol Cell, Vol. 13, 4279–95, (2002).
44. Kubista M, Rosner M, Kubista E, Bernaschek G, Hengstschlager M. “Brca1 regulates in vitro differentiation of mammary epithelial cells.” Oncogene, Vol. 21, 4747–56, (2002).
45. Soucek T, Holzl G, Bernaschek G, Hengstschlager M. “A role of the tuberous sclerosis gene-2 product in neuronal differentiation. ”, Oncogene, Vol. 16, 2197–204, (1998).
46. Prusa AR, Marton E, Rosner M, Bettelheim D, Lubec G, Pollak A, Bernaschek G, Hengstschlager M. “Neurogenic cells in human amniotic fluid. ” , Am J Obstet Gynecol, Vol. 191, 309–14, (2004)
47. Gronthos, S., Zannettino, A. C., Hay, S. J., Shi, S., Graves, S. E., Kortesidis, A., and Simmons, P. J.: “Molecular and cellular characterisation of highly purified stromal stem cells derived from human bone marrow”. J. Cell Sci., Vol. 116, 1827–1835, (2003).
48. Colter, D. C., Class, R., DiGirolamo, C.M., and Prockop, D. J.: “Rapid expansion of recycling stem cells in cultures of plastic-adherent cells from human bone marrow. ” Proc. Natl. Acad. Sci. USA, Vol. 97, 3213–3218, (2000).
49. Sekiya, I., Larson, B. L., Smith, J. R., Pochampally, R., Cui, J. G., and Prockop, D. J.: “Expansion of human adult stem cells from bone marrow stroma: conditions that maximize the yields of early progenitors and evaluate their quality.”Stem Cells, Vol. 20, 530–541, (2002).
50. Keisuke Okita, Tomoko Ichisaka & Shinya Yamanaka, “Generation of germline-competent induced pluripotent stem cells”, NATURE, Vol. 448, 313, (2007)
51. On line resources︰Sigma, http://www.sigma.com
52. On line resources︰BD, http://www.bdj.co.jp
53. Simmons, P. J. and Torok-Storb, B.: “Identification of stromal cell precursors in human bone marrow by a novel monoclonal antibody, STRO-1.”, Blood, Vol. 78, 55–62, (1991).
54. Gronthos, S., Graves, S. E., Ohta, S., and Simmons, P. J.: “The STRO-1+ fraction of adult human bone marrow contains the osteogenic precursors.” Blood, Vol. 84, 4164–4173, (1994).
55. Chi H. Lee, Anuj Singla, Yugyung Lee, “Biomedical applications of collagen”, International Journal of Pharmaceutics, Vol. 221, 1-22, (2001)
56. Huaping Tan, Jindan Wu, Lihong Lao, Changyou Gao, “Gelatin/chitosan/hyaluronan scaffold integrated with PLGA microspheres for cartilage tissue engineering”, Acta Biomaterialia, Vol. 5, 328-337, (2009)
57. N Mohan, PD Nair, Y tabata, “A 3D biodegradable protein base matrix for cartilage tissue engineering and stem cell differentiation to cartilage.”, J Mater Sci Master Med, 17, (2008)
58. Martin Ehrbar, Andrew Metters, Prisca Zammaretti, Jeffrey A. Hubbell, Andreas H. Zisch, “Endothelial cell proliferation and progenitor maturation by fibrin-bound VEGF variants with differential susceptibilities to local cellular activity”, Journal of Controlled Release, Vol. 101, 93-109, (2005)
59. S Cox, M Cole, B Tawil, “Behavior of human dermal fibroblasts in the three dimwnsional fibrin clots: dependwnce on fibrinogen anf thrombin concentration.”, Tissue Eng, Vol. 10, 942, (2004)
60. CC Tate, DA Shear, MC Tate et al. ,”Laminin and fibronectin scaffolds enhance neural stem cell transplanted into the injured brain”, Tissue Eng Regan Med, Vol. 3, 208,(2009)
61. CC Tate, DA Shear, MC Tate et al. ,”Fibronectin promotes survival and migration of primary neural stem cell transplanted into the traumatically injured mouse brain”, Cell Transplantation, Vol. 11, 238,(2002)
62. Schwartz, R. E., Reyes, M., Koodie, L., Jiang, Y., Blackstad, M., Lund, T., Lenvik, T., Johnson, S., Hu, W. S., and Verfaillie, C. M.: Multipotent adult progenitor cells from bone marrow differentiate into functional hepatocyte-like cells. J. Clin. Invest., 109, 1291–1302 (2002).
63. N Rangappa, A Romero, KD Nelson, “Laminin-coated poly(L-lactide) filaments induce robust neurite growth while providing directional orientation.”, J Biomed Master Res, Vol. 51, 625, (2000)
64. Williams, R. L., Hilton, D. J., Pease, S., Willson, T. A., Stewart, C. L., Gearing, D. P., Wagner, E. F., Metcalf, D., Nicola, N. A., and Gough, N. M.: Myeloid leukaemia inhibitory factor maintains the developmental potential of embryonic stem cells. Nature, 336, 684–687 (1988).
65. Smith, A.G., Heath, J. K., Donaldson, D. D., Wong, G.G., Moreau, J., Stahl, M., and Rogers, D.: Inhibition of pluripotential embryonic stem cell differentiation by purified polypeptides. Nature, 336, 688–690 (1988).
66. Amit, M., Carpenter, M. K., Inokuma, M. S., Chiu, C. P., Harris, C. P., Waknitz, M. A., Itskovitz-Eldor, J., and Thomson, J. A.: Clonally derived human embryonic stem cell lines maintain pluripotency and proliferative potential for prolonged periods of culture. Dev. Biol., 227, 271–278, (2000).
67. Judith M. Curran, Rui Chen, John A. Hunt, “The guidance of human mesenchymal stem cell differentiation in vitro by controlled modifications to the cell substrate”, Biomaterials, Vol. 27, 4783–4793, (2006).
68. Jennifer E. Phillips, Timothy A. Petrie, Francis P. Creighton, Andres J. Garcia, “Human mesenchymal stem cell differentiation on self-assembled monolayers presenting different surface chemistries”, Acta Biomaterialia, Vol. 6, 12-20, ( 2010)
69. Lichuan Qiana, W. Mark Saltzman, “Improving the expansion and neuronal differentiation of mesenchymal stem cells through culture surface modification”, Biomaterials, Vol. 25, 1331–1337, (2004)
70. Hongli Sun, Kai Feng, Jiang Hu, Shay Soker, Anthony Atala, Peter X. Ma, “Osteogenic differentiation of human amniotic fluid-derived stem cells induced by bone morphogenetic protein-7 and enhanced by nanofibrous scaffolds”, Biomaterials, Vol. 31, 1133–1139, (2010)
71. Ste’phane Roche, Marie-Jeanne Richard, and Marie-Christine Favrot, “Oct-4, Rex-1, and Gata-4 expression in human MSC increase the differentiation efficiency but not hTERT expression”, Journal of Cellular Biochemistry, Vol. 101, 271–280, (2007)
72. Caroline Seebach , Judith Schultheiss, Kerstin Wilhelm, Johannes Frank, Dirk Henrich, “Comparison of six bone-graft substitutes regarding to cell seeding efficiency, metabolism and growth behaviour of human mesenchymal stem cells (MSC) in vitro”, Injury, Int. J. Care Injured, (2010)
73. Jun-ichiro Jo, Ichio Aoki, Yasuhiko Tabata, “Design of iron oxide nanoparticles with different sizes and surface charges for simple and efficient labeling of mesenchymal stem cells”, Journal of Controlled Release, Vol. 142, 465–473, (2010)
74. Dave L. Roelen, Barbara J. van der Mast, Pieternella S. in’t Anker, Carin Kleijburg, Michael Eikmans, Els van Beelen, Godelieve M.J.S. de Groot-Swings, Wim E. Fibbe, Humphrey H.H. Kanhai, Sicco A. Scherjon, Frans H.J. Claas, “Differential immunomodulatory effects of fetal versus maternal multipotent stromal cells.”, Human Immunology, Vol. 70, 16-23, (2009)
75. Feng Zhao, Warren L. Grayson, Teng Ma, Bruce Bunnell, William W. Lu, “Effects of hydroxyapatite in 3-D chitosan–gelatin polymer network on human mesenchymal stem cell construct development”, Biomaterials, Vol. 27, 1859–1867,(2006)
76. A. J. Dulgar-Tulloch,1,2* R. Bizios,2,3** R. W. Siegel, “Human mesenchymal stem cell adhesion and proliferation in response to ceramic chemistry and nanoscale topography”, Journal of Biomedical Materials Research Part A, Vol. 90A, 586–594, (2009)
77. Lichuan Qiana, W. Mark Saltzman, “Improving the expansion and neuronal differentiation of mesenchymal stem cells through culture surface modification”, Biomaterials, Vol. 25, 1331–1337, (2004)
78. Jun-ichiro Jo, Ichio Aoki, Yasuhiko Tabata, “Design of iron oxide nanoparticles with different sizes and surface charges for simple and efficient labeling of mesenchymal stem cells”, Journal of Controlled Release, Vol. 142, 465–473, (2010)
79. Beckmen lecture
80. Y.W. Chung, Practical Guide to Surface Science and Spectroscopy, Academic Press, San Diego, (2001)
81. Xu, R.H., Peck, R. M., Li, D. S., Feng, X., Ludwig, T., and Thomson, J. A.: “Basic FGF and suppression of BMP signaling sustain undifferentiated proliferation of human ES cells. ” Nat. Methods, 2, 185–190 (2005).
82. Martin, I., Muraglia, A., Campanile, G., Cancedda, R., and Quarto, R.: “Fibroblast growth factor-2 supports ex vivo expansion and maintenance of osteogenic precursors from human bone marrow. ” Endocrinology, 138, 4456–4462 (1997).
83. K. Siegbahn, et al., ESCA: Atomic, Molecular and Solid State Structure Studied by means of Electron Spectroscopy, Almquist & Wiksells, Uppsala, (1967)
84. G. Ertl, J. K‥uppers, Low Energy Electrons and Surface Chemistry, VCH, Weinheim, (1985).
85. D. Briggs, M.P. Seah (Eds.), Practical Surface Analysis, Wiley, Chichester, (1990).
86. D.J. O’Connor, B.A. Sexton, R.St.C. Smart (Eds.), Surface Analysis Methods in Materiais Science, Springer-Verlag, Berlin, (1992).
87. Chadwick, K., Wang, L., Li, L., Menendez, P., Murdoch, B., Rouleau, A., and Bhatia, M.: “Cytokines and BMP-4 promote hematopoietic differentiation of human embryonic stem cells. ” , Blood, 102, 906–915, (2003).
88. On line resources︰A scientific and Industrial Research Organisation.
http://www.aaranyak.org/Projects/PCR.htm
89. Chien A, Edgar DB, and Trela JM, “Deoxyribonucleic acid polymerase from the extreme thermophile Thermus aquaticus”, J. Bacteriol, Vol 174, 1550–1557, (1976).
90. Lawyer FC, Stoffel S, Saiki RK, Chang SY, Landre PA, Abramson RD, and Gelfand DH “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. Vol 2, 275–287, (1993).
91. Freeman,W.M., Walker,S.J. and Vrana,K.E. “Quantitative RT-PCR: pitfalls and potential”, Biotechniques, Vol 26, 112–115, (1999).
92. 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 Res., Vol 17, 9437- 9446, (1989).
93. Gilliland, G., S. Perrin, K. Blanchard and H.F. Bunn., “Analysis of cytokine mRNA and DNA: detection and quantitation by competitive polymerase chain reaction”, Proc. Natl. Acad. Sci. USA, Vol 87, 2725-2729, (1990).
94. Rappolee, D.A., D. Mark, M.J. Banda and Z. Werb. “Wound macrophages express TGF-alpha and other growth factors in vivo: analysis by mRNA phenotyping”, Science, Vol 241, 708-712, (1988).
95. Ririe, K.M., R.P. Rasmussen and C.T. Wittwer., “Product differentiation by analysis of DNA melting curves during the polymerase chain reaction”, Anal. Biochem., Vol 245, 154-160, (1997).
96. Zhang, S. C., Wernig, M., Duncan, I. D., Brustle, O., and Thomson, J. A.: In vitro differentiation of transplantable neural precursors from human embryonic stem cells. Nat. Biotechnol., 19, 1129–1133, (2001).
97. Cheng S, Fockler C, Barnes WM, and Higuchi R, “Effective amplification of long targets from cloned inserts and human genomic DNA”, Proc Natl Acad Sci. Vol 91, 5695–5699, (1994).
98. Holland, P.M., R.D. Abramson, R. Watson and D.H. Gelfand. “Detection of specific polymerase chain reaction product by utilizing the 5’-3’exonuclease activity of Thermus aquaticus DNA polymerase”, Proc. Natl. Acad. Sci. USA, Vol 88, 7276-7280, (1991).
99. Wittwer, C.T., K.M. Ririe and R.P. Rasmussen., “Fluorescence monitoring of rapid cycle PCR for quantification”, In F. Ferre (Ed.), Gene Quantification. Birkhauser, Boston. , 129-144, (1998).
100. L Gucciardo,a R Lories,b N Ochsenbein-Ko‥ lble,a,* E Done’,a A Zwijsen,c J Deprest, “Fetal mesenchymal stem cells: isolation, properties and potential use in perinatology and regenerative medicine”, An International Journal of Obstetrics and Gynaecology
101. Jin HJ, Chen JS, Karageorgiou V et al. Human bone marrow stromal cell responses on electrospun silk fibroin mats. Biomaterials 2004;25:1039–1047.
102. Katz BZ, Zamir E, Bershadsky A et al. Physical state of the extracellular matrix regulates the structure and molecular composition of cell-matrix adhesions. Mol Biol Cell 2000;11:1047–1060.
103. Wang YK, Wang YH, Wang CZ et al. Rigidity of collagen fibrils controls collagen gel-induced down-regulation of focal adhesion complex proteins mediated by alpha2beta1 integrin. J Biol Chem 2003;278:21886–21892
104. Teratani, T., Yamamoto, H., Aoyagi, K., Sasaki, H., Asari, A., Quinn, G., Terada, M., and Ochiya, T. “Direct hepatic fate specification from mouse embryonic stem cells. ”Hepatology, 41, 836–846 (2005).
105. Farshid Guilak, Daniel M. Cohen, Bradley T. Estes,1 Jeffrey M. Gimble, Wolfgang Liedtke,and Christopher S. Chen, “Control of Stem Cell Fate by Physical Interactions with the Extracellular Matrix”, Cell Stem Cell, Vol. 5, (2009)
106. Peter X. Ma, “Biomimetic materials for tissue engineering.”, Advanced Drug Delivery Reviews, Vol. 60, 184–198 , (2008)
107. Heungsoo Shin, “Fabrication methods of an engineered microenvironment for analysis of cell–biomaterial interactions”, Biomaterials, Vol. 28, 126–133,(2007)
108. Unger RE, Huang Q, Peters K, Protzer D, Paul D, Kirkpatrick CJ. Growth of human cells on polyethersulfone (PES) hollow fiber membranes. Biomaterials 2005;26(14):1877–84.
109. Kinasiewicz A, Dudzinski K, Chwojnowski A, Werynski A, Kawiak J. Three-dimensional culture of hepatocytes on spongy polyethersulfone membrane developed for cell transplantation. Transplant Proc 2007;39(9):2914–6.
110. Wang T, Wang Y-q, Su Y-l, Jiang Z-y. Improved protein-adsorptionresistant property of PES/SPC blend membrane by adjustment of coagulation bath composition. Colloids Surf B Biointerfaces 2005;46(4):233–9.
111. Unger RE, Peters K, Huang Q, Funk A, Paul D, Kirkpatrick CJ. Vascularization and gene regulation of human endothelial cells growing on porous polyethersulfone (PES) hollow fiber membranes. Biomaterials 2005;26(17):3461–9.
112. Liu SX, Kim JT, Kim S. Effect of polymer surface modification onpolymer–protein interaction via hydrophilic polymer grafting. J Food Sci 2008;73(3):E143–50.
113. Ho MH et al. Promotion of cell affinity of porous PLLA scaffolds by immobilization of RGD peptides via plasma treatment. Macromol Biosci 2006;6(1):90–8.
114. Yang XB et al. Human osteoprogenitor growth and differentiation on synthetic biodegradable structures after surface modification. Bone 2001;29(6):523–31.
115. Senyah N, Hildebrand G, Liefeith K. Comparison between RGD peptide-modified titanium and borosilicate surfaces. Anal Bioanal Chem 2005;383:758–62
116. Santiago LY, Nowak RW, Peter Rubin J, Marra KG. Peptide-surface modification of poly(caprolactone) with laminin-derived sequences for adipose-derived stem cell applications. Biomaterials 2006;27(15): 2962–9.
117. J.L.West, J.A. Hubbell, “Polymeric biomaterials with degradation sites for proteases involved in cell migration”, Macromolecules, Vol. 32 241–244, (1999).,
118. B.C. Isenberg, C. Williams, R.T. Tranquillo, “Small-diameter artificial arteries engineered in vitro”, Circ. Res., Vol. 98, 25–35, (2006).
119. C.G. Pitt, M.M. Gratzl, G.L. Kimmel, J. Surles, A. Schindler, Aliphatic polyesters II. “The degradation of poly (DL-lactide), poly (epsiloncaprolactone), and their copolymers in vivo”, Biomaterials, Vol. 2, 215–220, (1981).
120. S.C. Woodward, P.S. Brewer, F. Moatamed, A. Schindler, C.G. Pitt, “The intracellular degradation of poly(epsilon-caprolactone)”, J. Biomed. Mater. Res., Vol. 19, 437–444, (1985).
121. P.X. Ma, Tissue engineering, in: J.I. Kroschwitz (Ed.), Third ed., “Encyclopedia of Polymer Science and Technology”, JohnWiley & Sons, Inc., Hoboken, NJ, vol. 12, 261–291, (2005).
122. E.R. Wright, V.P. Conticello, “Self-assembly of block copolymers derived from elastin-mimetic polypeptide sequences”, Adv. Drug Deliv. Rev., Vol. 54, 1057–1073, (2002).
123. H. Betre, S.R. Ong, F. Guilak, A. Chilkoti, B. Fermor, L.A. Setton, “Chondrocytic differentiation of human adipose-derived adult stem cells in elastin-like polypeptide”, Biomaterials, Vol. 27, 91–99, (2006).
124. Y. Cao, H. Li, “Polyprotein of GB1 is an ideal artificial elastomeric protein”, Nat. Matters, Vol. 6, 109–114, (2007).
125. E.D. Hay, “Cell biology of extracellular matrix”. 2nd ed. Vol., New York: Plenum Press, (1991).
126. T. Elsdale, J. Bard, “Collagen substrata for studies on cell behavior”, J. Cell Biol., Vol. 54, 626–637, (1972).
127. W.J. Morton, “Method of dispersing fluids”. US patent, Vol. 705, 691, (1902).
128. J.A. Matthews, G.E. Wnek, D.G. Simpson, G.L. Bowlin, “Electrospinning of collagen nanofibers”, Biomacromolecules, Vol. 3, 232–238, (2002).
129. W.J. Li, C.T. Laurencin, E.J. Caterson, R.S. Tuan, F.K. Ko, “Electrospun nanofibrous structure: a novel scaffold for tissue engineering”, J. Biomed. Mater. Res., Vol. 60, 613–621, (2002).
130. F. Yang, R. Murugan, S.Wang, S. Ramakrishna, “Electrospinning of nano/micro scale poly(L-lactic acid) aligned fibers and their potential in neural tissue engineering”, Biomaterials, Vol. 26, 2603–2610, (2005).
131. K.N. Chua, W.S. Lim, P. Zhang, H. Lu, J. Wen, S. Ramakrishna, K. W.Leong, H.Q. Mao, “Stable immobilization of rat hepatocyte spheroids on galactosylated nanofiber scaffold”, Biomaterials, Vol. 26, 2537–2547, (2005).
132. W.-j. Li, K.G. Danielson, P.G. Alexander, R.S. Tuan, “Biological response of chondrocytes cultured in three-dimensional nanofibrous poly(ecaprolactone) scaffolds”, J. Biomed. Mater. Res., Part A, Vol. 67A, 1105–1114, (2003).
133. Q.P. Pham, U. Sharma, A.G. Mikos, “Electrospun poly(epsilon-caprolactone) microfiber and multilayer nanofiber/microfiber scaffolds: characterization of scaffolds and measurement of cellular infiltration”, Biomacromolecules, Vol. 7, 2796–2805, (2006).
指導教授 樋口亞紺(Akon Higuchi) 審核日期 2010-7-15
推文 facebook   plurk   twitter   funp   google   live   udn   HD   myshare   reddit   netvibes   friend   youpush   delicious   baidu   
網路書籤 Google bookmarks   del.icio.us   hemidemi   myshare   

若有論文相關問題,請聯絡國立中央大學圖書館推廣服務組 TEL:(03)422-7151轉57407,或E-mail聯絡  - 隱私權政策聲明