博碩士論文 106324013 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:159 、訪客IP:3.145.60.29
姓名 李雨駿(Yu-Chun Lee)  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 膜遷移法藉由細胞外間質塗覆薄膜從脂肪組織分離出脂肪幹細胞
(A Hybrid-Membrane Migration Method to Isolate Adipose-Derived Stem Cells from Fat Tissues Through Membranes Coated with Extracellular Matrices)
相關論文
★ 於不同彈性係數的生醫材料上體外培植造血幹細胞★ 藉由調整水凝膠之表面電荷及軟硬度並嫁接玻連蛋白用以培養人類多功能幹細胞
★ 可見光對羊水間葉幹細胞成骨分化之影響★ 可見光調控神經細胞之基因表現及突觸生長
★ 膜純化法及免疫抗體磁珠法用於分離及體外增殖血液幹細胞之研究★ 人類表皮成長因子的結構穩定性及生物活性測定
★ 微環境對羊水間葉幹細胞多功能性基因表現及分化之影響★ 奈米片段與細胞外基質之改質膜用於臍帶血中造血幹細胞之純化與培養
★ 小鼠脂肪幹細胞之膜純化法及細胞外間質對人類脂肪幹細胞影響之研究★ 利用具有奈米片段與細胞外間質蛋白質的表面改殖材質進行臍帶血造血幹細胞體外培養
★ 在不同培養條件下針對大腸癌細胞及組織中癌細胞進行純化、剔除及鑑定之研究★ 羊水間葉幹細胞培養於細胞外間質改質表面其分化能力及多能性之研究
★ 人類脂肪幹細胞的膜純化法與分化能力研究★ 具有抗藥性之大腸癌細胞株能提高癌胚抗原的表現,但並非是癌症起始細胞
★ 羊水間葉幹細胞培養於接枝細胞外間質寡肽與環狀肽具有最佳表面硬度的生醫材料,其增殖能力及多能性之研究★ 人類體細胞從組成誘導型多能性幹細胞培養在無飼養層上
檔案 [Endnote RIS 格式]    [Bibtex 格式]    [相關文章]   [文章引用]   [完整記錄]   [館藏目錄]   [檢視]  [下載]
  1. 本電子論文使用權限為同意立即開放。
  2. 已達開放權限電子全文僅授權使用者為學術研究之目的,進行個人非營利性質之檢索、閱讀、列印。
  3. 請遵守中華民國著作權法之相關規定,切勿任意重製、散佈、改作、轉貼、播送,以免觸法。

摘要(中) 人體脂肪幹細胞(hADSCs)可以通過從脂肪組織中分離獲得,且hADSCs目前是比人體誘導多能幹細胞(hiPSC)和胚胎幹細胞(hESC)更實用的幹細胞來源。目前,多數的臨床試驗使用hADSCs,而僅使用hiPSC和hESC進行了少數臨床試驗。已知hADSCs顯示異質性特徵並且包含不同的多能性和分化能力。因此,我們預期通過不同分離方式分離的hADSCs,其幹細胞特徵,多能性和分化能力應該有所不同。普遍利用基質血管部分(SVF,原生hADSCs溶液)的細胞培養來得到hADSCs,其中SVF溶液可通過膠原酶(Collagenase)消化脂肪組織並且離心後獲得。依不同純化方式所分離出的人類脂肪幹細胞會具有不同的純度及多樣性。在實驗室,我將通過網狀尼龍膜過濾器的膜遷移方法進行創新,脂肪組織溶液經由此方法,使其hADSCs純化且具有極高的純度和多能性。其方法將原生脂肪組織溶液通過孔徑為8至25μm的多孔膜(例如:尼龍網),並將膜放入細胞培養液中培養15-18天。
就我們所知,其生長的微環境於人類脂肪幹細胞之基因表現及分化能力中,扮演著重要的角色。因此在此研究中,新的膜遷移方法是使用最佳孔徑(11和20μm)的尼龍網膜和PLGA絲網膜開發的,其中最佳細胞外間質(ECM)塗覆在膜上,此方法可以純化hADSCs。期望利用優化後的純化方法所獲得之脂肪幹細胞能具有較高多能性,以及細胞分化能力,如:軟骨細胞、成骨細胞及脂肪細胞之分化。我們利用膜遷移方法將脂肪組織分離出hADSCs,並且在此方法中使用了不同的膜,如:(a)尼龍網(孔徑為11跟20μm)和PLGA絲網膜、(b)尼龍網(孔徑為11跟20μm)和PLGA絲網膜塗佈第一型膠原蛋白(Collagen type I)、(c)尼龍網(孔徑為11跟20μm)和PLGA絲網膜塗佈人類纖維連接蛋白(Fibronectin)、(d)尼龍網(孔徑為11跟20μm)和PLGA絲網膜塗佈人類重組玻璃粘連蛋白(human recombinant Vitronectin)。第一型膠原蛋白為異種來源的材料,其餘細胞外間質之來源則為人類(無異種材料)。從膜遷移下來的人體脂肪幹細胞(hADSCs)與使用普通培養方法分離的細胞相比較表現極高百分比(例如:> 98%)的間充質幹細胞標記物(CD44,CD73和CD90),並且在一些多能性基因(Oct4,Sox2和Nanog)的表現量高出一個級距。
摘要(英) Human adipose-derived stem cells, hADSCs, can be obtained by isolation from fat tissue, which is currently a more practical source of stem cells than human induced pluripotent stem cells (hiPSCs) and embryonic stem cells (hESCs). Currently, several clinical trials use hADSCs, whereas only a few clinical trials have been performed using hiPSCs and hESCs. However, hADSCs are known to show heterogeneous characteristics and contain different pluripotency and differentiation abilities. Therefore, it is expected that the stem cell characteristics, pluripotency, and differentiation abilities should be different for hADSCs isolated by different isolation methods. hADSCs are typically isolated by cell culture of stromal vascular fraction (SVF, primary hADSC solution) where the SVF solution can be obtained by collagenase digestion of fat tissues followed by centrifugation. The isolated hADSCs can possess different purity levels and divergent properties depending on the purification methods used. It is innovated that the membrane migration method through Nylon mesh filter purifies hADSCs from a fat tissue solution with extremely high purity and pluripotency in my laboratory. A primary fat-tissue solution was permeated through the porous membranes (e.g., Nylon mesh) with a pore size from 8 to 25 μm, and the membranes were incubated in cell culture medium for 15-18 days.
In this study, I developrd a new membrane migration method using Nylon mesh membranes having optimal pore sizes, 11 and 20 μm, and PLGA/silk membranes where optimal extracellular matrix (ECM) was coated on the membranes, which could purify hADSCs. The isolated hADSCs are expected to have high pluripotency and high differentiation ability into chondrocytes, osteoblasts and adipocytes. hADSCs were isolated from adipose tissue by the membrane migration method where different membranes were used, e.g., (a) Nylon mesh and PLGA (poly (lactic-co-glycolic acid))/silk screen membrane, (b) Nylon mesh and PLGA/silk screen membrane coated with collagen type I, (c) Nylon mesh and PLGA/silk screen membrane coated with human recombinant-vitronectin, (d) Nylon mesh and PLGA/silk screen membrane coated with human fibronectin. Collagen type I is xeno-containing materials, whereas another extracellular matrices (ECMs) were xeno-free materials. The hADSCs that migrated from the membranes kept an extremely high percentage (e.g., >98%) expression of mesenchymal stem cell markers (CD44, CD73, and CD90) and showed almost one order of magnitude higher expression of some pluripotency genes (Oct4, Sox2, and Nanog) compared with cells isolated using the conventional culture method.
關鍵字(中) ★ 脂肪幹細胞
★ 細胞外間質
★ 膜分離法
關鍵字(英) ★ Adipose derived stem cell
★ Extracellular matrix
★ membrane migration method
論文目次 Abstract i
Index of Content iii
Index of Figure vii
Index of Table xi
Chapter 1. Introduction 1
1-1 Stem cell therapies in clinical trials 1
1-1.1 Stem cells 2
1-1.2 Pluripotent stem cells (PSCs) 4
1-1.3 Mesenchymal stem cells (MSCs) 5
1-2 Cell migration 7
1-3 Adipose derived-stem cells (ADSCs) 8
1-3.1 Isolation of adipose derived-stem cells 9
1-3.2 Surface immunophenotype marker of ADSCs 10
1-3.3 Purification of membrane filtration and migration method 13
1-3.4 Differentiation abilities of adipose derived-stem cells 14
1-4 Extracellular matrix (ECM) 17
1-4.1 Collagen (Col) 18
1-4.2 Fibronectin (FN) 20
1-4.3 Vitronectin (VN) 21
1-5 Poly (lactic-co-glycolic acid) (PLGA)/silk screen membrane 22
1-6 Goal of this study 23
Chapter 2 Materials and Methods 25
2-1 Experimental materials 25
i. Adipose tissue 25
ii. Common cell cultured dish 26
iii. Isolation process 26
iv. Membrane migration method 26
v. Isolation process 27
vi. Passage process 27
vii. pH standard solution 27
viii. Phosphate buffer saline solution (PBS) 27
ix. Flow cytometry 28
x. RNA extraction kit 28
xi. Reverse transcription kit 28
xii. Real-time polymerization chain reaction 28
xiii. qRT-PCR probe 29
xiv. Osteogenic differentiation 29
xv. Chondrogenic differentiation 30
xvi. Adipogenic differentiation 30
2-2 Experimental methods 31
2-2.1 Preparation of phosphate buffer solution (PBS) 31
2-2.2 Preparation of culture medium 31
2-2.3 Isolation of adipose-derived stem cells (hADSCs) 31
2-2.4 Cultivation and passage of hADSCs 33
2-2.5 Cell density measurement 35
2-2.6 Preparation of PLGA/silk screen membrane 36
2-2.7 Preparation of ECM-coated membranes 37
2-2.8 Pluripotent gene expression analysis 38
2-3.9 Immunofluorescence staining 42
2-3.10 Flow-cytometry measurements 43
2-3.11 Quantitative analysis of differentiation 44
2-3.12 Osteogenic differentiation 45
2-3.13 Alkaline phosphate activity (ALP activity) 45
2-3.14 Alizarin red S staining assay 47
2-3.15 von Kossa staining 47
2-3.16 Adipogenic differentiation of hADSCs 48
2-3.17 Oil Red O staining 48
2-3.18 Chondrogenic differentiation of hADSCs 48
2-3.19 Alcian blue staining 49
Chapter 3 Results and Discussion 50
3-1 Structure of Nylon and PLGA/silk screen membranes 50
3-2 Cultivation of hADSCs 55
3-2.1 Morphology and doubling time of passage 5 hADSCs after membrane migration method 56
3-2.2 The cell ratio of hADSCs (passage 5) by membrane filtration and migration method 60
3-3 Differentiation abilities of hADSCs 62
3-3.1 Osteogenic differentiation of isolated hADSCs (cell line, passage 5) by membrane migration method 63
3-3.2 Osteogenic differentiation of isolated hADSCs (Primary fat 83
3-3.3 Chondrogenic differentiation of isolated hADSCs (cell line, 92
3-3.4 Adipogenic differentiation of isolated hADSCs (cell line, 96
Chapter 4 Conclusions 98
Reference 101
Supplementary data 111
S-1 . Osteogenic differentiation of isolated hADSCs (cell line, passage 5) 113
S-2 . Osteogenic differentiation of isolated hADSCs (Primary) 121
S-3 . Adipogenic differentiation of isolated hADSCs (cell line, passage 5) 129
參考文獻 [1] J. K. Biehl and B. Russell, "Introduction to Stem Cell Therapy," (in English), J. Cardiovasc. Nurs., Article vol. 24, no. 2, pp. 98-103, Mar-Apr 2009.
[2] H. Mizuno, M. Tobita, and A. C. Uysal, "Concise Review: Adipose-Derived Stem Cells as a Novel Tool for Future Regenerative Medicine," (in English), Stem Cells, Review vol. 30, no. 5, pp. 804-810, May 2012.
[3] P. Marks and S. Gottlieb, "Balancing Safety and Innovation for Cell-Based Regenerative Medicine," (in English), N. Engl. J. Med., Article vol. 378, no. 10, pp. 954-959, Mar 2018.
[4] G. Cossu et al., "Lancet Commission: Stem cells and regenerative medicine," (in English), Lancet, Review vol. 391, no. 10123, pp. 883-910, Mar 2018.
[5] R. Duelen and M. Sampaolesi, "Stem cell technology in cardiac regeneration: a pluripotent stem cell promise," EBioMedicine, vol. 16, pp. 30-40, 2017.
[6] A. Trounson and C. McDonald, "Stem Cell Therapies in Clinical Trials: Progress and Challenges," (in English), Cell Stem Cell, Review vol. 17, no. 1, pp. 11-22, Jul 2015.
[7] G. Kolios and Y. Moodley, "Introduction to stem cells and regenerative medicine," Respiration, vol. 85, no. 1, pp. 3-10, 2013.
[8] P. A. Zuk et al., "Human adipose tissue is a source of multipotent stem cells," (in English), Mol. Biol. Cell, Article vol. 13, no. 12, pp. 4279-4295, Dec 2002.
[9] B. A. Bunnell, M. Flaat, C. Gagliardi, B. Patel, and C. Ripoll, "Adipose-derived stem cells: Isolation, expansion and differentiation," (in English), Methods, Article vol. 45, no. 2, pp. 115-120, Jun 2008.
[10] P. R. Baraniak and T. C. McDevitt, "Stem cell paracrine actions and tissue regeneration," (in English), Regen. Med., Review vol. 5, no. 1, pp. 121-143, Jan 2010.
[11] S. W. Lane, D. A. Williams, and F. M. Watt, "Modulating the stem cell niche for tissue regeneration," (in English), Nat. Biotechnol., Article vol. 32, no. 8, pp. 795-803, Aug 2014.
[12] V. Volarevic et al., "Ethical and Safety Issues of Stem Cell-Based Therapy," (in English), Int. J. Med. Sci., Review vol. 15, no. 1, pp. 36-45, 2018.
[13] J. M. Gimble, A. J. Katz, and B. A. Bunnell, "Adipose-derived stem cells for regenerative medicine," (in English), Circ.Res., Review vol. 100, no. 9, pp. 1249-1260, May 2007.
[14] C. L. Baker and M. F. Pera, "Capturing totipotent stem cells," Cell Stem Cell, vol. 22, no. 1, pp. 25-34, 2018.
[15] K. Takahashi and S. Yamanaka, "Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors," cell, vol. 126, no. 4, pp. 663-676, 2006.
[16] M. Dominici et al., "Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement," Cytotherapy, vol. 8, no. 4, pp. 315-317, 2006.
[17] M. F. Pittenger et al., "Multilineage potential of adult human mesenchymal stem cells," science, vol. 284, no. 5411, pp. 143-147, 1999.
[18] I. J. Fox, G. Q. Daley, S. A. Goldman, J. Huard, T. J. Kamp, and M. Trucco, "Use of differentiated pluripotent stem cells in replacement therapy for treating disease," (in English), Science, Review vol. 345, no. 6199, pp. 889-+, Aug 2014, Art no. Unsp 1247391.
[19] P. P. Hou et al., "Pluripotent Stem Cells Induced from Mouse Somatic Cells by Small-Molecule Compounds," (in English), Science, Article vol. 341, no. 6146, pp. 651-654, Aug 2013.
[20] R. Sugimura et al., "Haematopoietic stem and progenitor cells from human pluripotent stem cells," (in English), Nature, Article vol. 545, no. 7655, pp. 432-+, May 2017.
[21] R. J. Thomas et al., "Automated, Scalable Culture of Human Embryonic Stem Cells in Feeder-Free Conditions," (in English), Biotechnol. Bioeng., Article vol. 102, no. 6, pp. 1636-1644, Apr 2009.
[22] M. F. Pera, B. Reubinoff, and A. Trounson, "Human embryonic stem cells," (in English), J. Cell Sci., Editorial Material vol. 113, no. 1, pp. 5-10, Jan 2000.
[23] I. de Melo-Martin, Z. Rosenwaks, and J. J. Fins, "New methods for deriving embryonic stem cell lines: are the ethical problems solved?," (in English), Fertil. Steril., Article vol. 86, no. 5, pp. 1330-1332, Nov 2006.
[24] Y. H. Loh et al., "Generation of induced pluripotent stem cells from human blood," (in English), Blood, Article vol. 113, no. 22, pp. 5476-5479, May 2009.
[25] A. I. Caplan, "Mesenchymal stem cells," Journal of orthopaedic research, vol. 9, no. 5, pp. 641-650, 1991.
[26] X. Wei, X. Yang, Z. P. Han, F. F. Qu, L. Shao, and Y. F. Shi, "Mesenchymal stem cells: a new trend for cell therapy," (in English), Acta Pharmacol. Sin., Review vol. 34, no. 6, pp. 747-754, Jun 2013.
[27] D. Levy, T. C. De Melo, J. L. Ruiz, and S. P. Bydlowski, "Oxysterols and mesenchymal stem cell biology," Chemistry and physics of lipids, vol. 207, pp. 223-230, 2017.
[28] M. Dominici et al., "Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement," (in English), Cytotherapy, Article vol. 8, no. 4, pp. 315-317, Aug 2006.
[29] T. Yi and S. U. Song, "Immunomodulatory Properties of Mesenchymal Stem Cells and Their Therapeutic Applications," (in English), Arch. Pharm. Res., Review vol. 35, no. 2, pp. 213-221, Feb 2012.
[30] C. Nesselmann et al., "Mesenchymal stem cells and cardiac repair," (in English), J. Cell. Mol. Med., Review vol. 12, no. 5B, pp. 1795-1810, Oct 2008.
[31] K. Shah, "Mesenchymal stem cells engineered for cancer therapy," (in English), Adv. Drug Deliv. Rev., Review vol. 64, no. 8, pp. 739-748, Jun 2012.
[32] K. LeBlanc et al., "Mesenchymal stem cells for treatment of steroid-resistant, severe, acute graft-versus-host disease: a phase II study," (in English), Lancet, Article vol. 371, no. 9624, pp. 1579-1586, May 2008.
[33] V. Karantalis and J. M. Hare, "Use of Mesenchymal Stem Cells for Therapy of Cardiac Disease," (in English), Circ.Res., Review vol. 116, no. 8, pp. 1413-1430, Apr 2015.
[34] G. Chamberlain, J. Fox, B. Ashton, and J. Middleton, "Concise review: Mesenchymal stem cells: Their phenotype, differentiation capacity, immunological features, and potential for homing," (in English), Stem Cells, Review vol. 25, no. 11, pp. 2739-2749, Nov 2007.
[35] E. H. Barriga, K. Franze, G. Charras, and R. Mayor, "Tissue stiffening coordinates morphogenesis by triggering collective cell migration in vivo," (in eng), Nature, vol. 554, no. 7693, pp. 523-527, Feb 22 2018.
[36] L. Li, Y. He, M. Zhao, and J. Jiang, "Collective cell migration: Implications for wound healing and cancer invasion," (in eng), Burns & trauma, vol. 1, no. 1, pp. 21-6, 2013.
[37] A. J. Ridley et al., "Cell migration: integrating signals from front to back," (in eng), Science, vol. 302, no. 5651, pp. 1704-9, Dec 5 2003.
[38] Y. J. Liu et al., "Confinement and low adhesion induce fast amoeboid migration of slow mesenchymal cells," (in eng), Cell, vol. 160, no. 4, pp. 659-672, Feb 12 2015.
[39] K. Talkenberger, E. A. Cavalcanti-Adam, A. Voss-Bohme, and A. Deutsch, "Amoeboid-mesenchymal migration plasticity promotes invasion only in complex heterogeneous microenvironments," (in eng), Scientific reports, vol. 7, no. 1, p. 9237, Aug 23 2017.
[40] N. Kalinina et al., "Characterization of secretomes provides evidence for adipose-derived mesenchymal stromal cells subtypes," Stem cell research & therapy, vol. 6, no. 1, p. 221, 2015.
[41] R. Dai, Z. J. Wang, R. Samanipour, K. I. Koo, and K. Kim, "Adipose-Derived Stem Cells for Tissue Engineering and Regenerative Medicine Applications," (in English), Stem Cells Int., Review p. 19, 2016, Art no. 6737345.
[42] D. Y. Kim and J. H. Sung, "Regulatory role of microRNAs in the proliferation and differentiation of adipose-derived stem cells," (in English), Histol. Histopath., Review vol. 32, no. 1, pp. 1-10, Jan 2017.
[43] E. J. Combellack et al., "Adipose regeneration and implications for breast reconstruction: update and the future," (in English), Gland Surg., Review vol. 5, no. 2, pp. 227-241, Apr 2016.
[44] S. J. Huang et al., "Adipose-Derived Stem Cells: Isolation, Characterization, and Differentiation Potential," (in English), Cell Transplant., Review vol. 22, no. 4, pp. 701-709, 2013.
[45] S. L. Francis, S. Duchi, C. Onofrillo, C. Di Bella, and P. F. M. Choong, "Adipose-Derived Mesenchymal Stem Cells in the Use of Cartilage Tissue Engineering: The Need for a Rapid Isolation Procedure," (in English), Stem Cells Int., Review p. 9, 2018, Art no. 8947548.
[46] F. Ng et al., "PDGF, TGF-beta, and FGF signaling is important for differentiation and growth of mesenchymal stem cells (MSCs): transcriptional profiling can identify markers and signaling pathways important in differentiation of MSCs into adipogenic, chondrogenic, and osteogenic lineages," (in English), Blood, Article vol. 112, no. 2, pp. 295-307, Jul 2008.
[47] T. R. Nayak et al., "Graphene for Controlled and Accelerated Osteogenic Differentiation of Human Mesenchymal Stem Cells," (in English), ACS Nano, Article vol. 5, no. 6, pp. 4670-4678, Jun 2011.
[48] B. T. Estes, B. O. Diekman, J. M. Gimble, and F. Guilak, "Isolation of adipose-derived stem cells and their induction to a chondrogenic phenotype," (in English), Nat. Protoc., Article vol. 5, no. 7, pp. 1294-1311, 2010.
[49] R. Talens-Visconti et al., "Hepatogenic differentiation of human mesenchymal stem cells from adipose tissue in comparison with bone marrow mesenchymal stem cells," (in English), World J. Gastroenterol., Article vol. 12, no. 36, pp. 5834-5845, Sep 2006.
[50] J. L. Boulland et al., "Epigenetic Regulation of Nestin Expression During Neurogenic Differentiation of Adipose Tissue Stem Cells," (in English), Stem Cells Dev., Article vol. 22, no. 7, pp. 1042-1052, Apr 2013.
[51] K. Yoshimura et al., "Characterization of freshly isolated and cultured cells derived from the fatty and fluid portions of liposuction aspirates," (in English), J. Cell. Physiol., Article vol. 208, no. 1, pp. 64-76, Jul 2006.
[52] M. Rodbell and A. B. Jones, "Metabolism of isolated fat cells. 3. The similar inhibitory action of phospholipase C (Clostridium perfringens alpha toxin) and of insulin on lipolysis stimulated by lipolytic hormones and theophylline," (in eng), The Journal of biological chemistry, vol. 241, no. 1, pp. 140-2, Jan 10 1966.
[53] M. Rodbell, "Metabolism of isolated fat cells. II. The similar effects of phospholipase C (Clostridium perfringens alpha toxin) and of insulin on glucose and amino acid metabolism," (in eng), The Journal of biological chemistry, vol. 241, no. 1, pp. 130-9, Jan 10 1966.
[54] M. Rodbell, "The metabolism of isolated fat cells. IV. Regulation of release of protein by lipolytic hormones and insulin," (in eng), The Journal of biological chemistry, vol. 241, no. 17, pp. 3909-17, Sep 10 1966.
[55] R. L. Van, C. E. Bayliss, and D. A. Roncari, "Cytological and enzymological characterization of adult human adipocyte precursors in culture," (in eng), The Journal of clinical investigation, vol. 58, no. 3, pp. 699-704, Sep 1976.
[56] A. Bajek, N. Gurtowska, J. Olkowska, L. Kazmierski, M. Maj, and T. Drewa, "Adipose-Derived Stem Cells as a Tool in Cell-Based Therapies," (in English), Arch. Immunol. Ther. Exp., Review vol. 64, no. 6, pp. 443-454, Dec 2016.
[57] W. Q. Zhan, S. S. Tan, and F. Lu, "Adipose-Derived Stem Cell Delivery for Adipose Tissue Engineering: Current Status and Potential Applications in a Tissue Engineering Chamber Model," (in English), Stem Cell Rev. Rep., Review vol. 12, no. 4, pp. 484-491, Aug 2016.
[58] S. Gronthos, D. M. Franklin, H. A. Leddy, P. G. Robey, R. W. Storms, and J. M. Gimble, "Surface protein characterization of human adipose tissue-derived stromal cells," (in eng), J Cell Physiol, vol. 189, no. 1, pp. 54-63, Oct 2001.
[59] K. McIntosh et al., "The immunogenicity of human adipose-derived cells: Temporal changes in vitro," (in English), Stem Cells, Article vol. 24, no. 5, pp. 1246-1253, May 2006.
[60] J. B. Mitchell et al., "Immunophenotype of human adipose-derived cells: Temporal changes in stromal-associated and stem cell-associated markers," (in English), Stem Cells, Article vol. 24, no. 2, pp. 376-385, Feb 2006.
[61] D. C. Chen et al., "Purification of human adipose-derived stem cells from fat tissues using PLGA/silk screen hybrid membranes," (in eng), Biomaterials, vol. 35, no. 14, pp. 4278-87, May 2014.
[62] A. Higuchi et al., "Differentiation ability of adipose-derived stem cells separated from adipose tissue by a membrane filtration method," (in English), J. Membr. Sci., Article vol. 366, no. 1-2, pp. 286-294, Jan 2011.
[63] C. H. Wu et al., "The isolation and differentiation of human adipose-derived stem cells using membrane filtration," (in eng), Biomaterials, vol. 33, no. 33, pp. 8228-39, Nov 2012.
[64] A. Higuchi, Y. Shindo, Y. Gomei, T. Mori, T. Uyama, and A. Umezawa, "Cell separation between mesenchymal progenitor cells through porous polymeric membranes," (in eng), Journal of biomedical materials research. Part B, Applied biomaterials, vol. 74, no. 1, pp. 511-9, Jul 2005.
[65] H. R. Lin et al., "Purification and differentiation of human adipose-derived stem cells by membrane filtration and membrane migration methods," (in eng), Scientific reports, vol. 7, p. 40069, Jan 10 2017.
[66] A. Higuchi et al., "A hybrid-membrane migration method to isolate high-purity adipose-derived stem cells from fat tissues," (in eng), Scientific reports, vol. 5, p. 10217, May 13 2015.
[67] F. Guilak et al., "Clonal analysis of the differentiation potential of human adipose-derived adult stem cells," (in English), J. Cell. Physiol., Article vol. 206, no. 1, pp. 229-237, Jan 2006.
[68] P. A. Zuk et al., "Multilineage cells from human adipose tissue: implications for cell-based therapies," (in eng), Tissue Eng, vol. 7, no. 2, pp. 211-28, Apr 2001.
[69] G. Di Rocco et al., "Myogenic potential of adipose-tissue-derived cells," (in eng), J Cell Sci, vol. 119, no. Pt 14, pp. 2945-52, Jul 15 2006.
[70] M. Q. Wickham, G. R. Erickson, J. M. Gimble, T. P. Vail, and F. Guilak, "Multipotent stromal cells derived from the infrapatellar fat pad of the knee," (in eng), Clinical orthopaedics and related research, no. 412, pp. 196-212, Jul 2003.
[71] J. M. Gimble and F. Guilak, "Differentiation potential of adipose derived adult stem (ADAS) cells," (in eng), Current topics in developmental biology, vol. 58, pp. 137-60, 2003.
[72] S. S. Tholpady, R. Llull, R. C. Ogle, J. P. Rubin, J. W. Futrell, and A. J. Katz, "Adipose tissue: stem cells and beyond," (in eng), Clinics in plastic surgery, vol. 33, no. 1, pp. 55-62, vi, Jan 2006.
[73] A. Schaffler and C. Buchler, "Concise review: adipose tissue-derived stromal cells--basic and clinical implications for novel cell-based therapies," (in eng), Stem Cells, vol. 25, no. 4, pp. 818-27, Apr 2007.
[74] F. Haugen and C. A. Drevon, "The interplay between nutrients and the adipose tissue," (in eng), The Proceedings of the Nutrition Society, vol. 66, no. 2, pp. 171-82, May 2007.
[75] C. Contreras et al., "The brain and brown fat," (in eng), Annals of medicine, vol. 47, no. 2, pp. 150-68, Mar 2015.
[76] S. Karahuseyinoglu, C. Kocaefe, D. Balci, E. Erdemli, and A. Can, "Functional structure of adipocytes differentiated from human umbilical cord stroma-derived stem cells," (in eng), Stem Cells, vol. 26, no. 3, pp. 682-91, Mar 2008.
[77] F. Caviggioli, V. Vinci, A. Salval, and M. Klinger, "Human adipose-derived stem cells: isolation, characterization and applications in surgery," (in eng), ANZ journal of surgery, vol. 79, no. 11, p. 856, Nov 2009.
[78] R. K. Jaiswal, N. Jaiswal, S. P. Bruder, G. Mbalaviele, D. R. Marshak, and M. F. Pittenger, "Adult human mesenchymal stem cell differentiation to the osteogenic or adipogenic lineage is regulated by mitogen-activated protein kinase," (in English), J. Biol. Chem., Article vol. 275, no. 13, pp. 9645-9652, Mar 2000.
[79] T. Mochizuki et al., "Higher chondrogenic potential of fibrous synovium- and adipose synovium-derived cells compared with subcutaneous fat-derived cells: distinguishing properties of mesenchymal stem cells in humans," (in eng), Arthritis and rheumatism, vol. 54, no. 3, pp. 843-53, Mar 2006.
[80] J. S. Park et al., "The effect of matrix stiffness on the differentiation of mesenchymal stem cells in response to TGF-beta," (in English), Biomaterials, Article vol. 32, no. 16, pp. 3921-3930, Jun 2011.
[81] D. Bosnakovski, M. Mizuno, G. Kim, S. Takagi, M. Okumura, and T. Fujinaga, "Chondrogenic differentiation of bovine bone marrow mesenchymal stem cells (MSCs) in different hydrogels: Influence of collagen type II extracellular matrix on MSC chondrogenesis," (in English), Biotechnol. Bioeng., Article vol. 93, no. 6, pp. 1152-1163, Apr 2006.
[82] A. M. Mackay, S. C. Beck, J. M. Murphy, F. P. Barry, C. O. Chichester, and M. F. Pittenger, "Chondrogenic differentiation of cultured human mesenchymal stem cells from marrow," (in English), Tissue Eng., Article vol. 4, no. 4, pp. 415-428, Win 1998.
[83] P. Singh and J. E. Schwarzbauer, "Fibronectin and stem cell differentiation - lessons from chondrogenesis," (in eng), J Cell Sci, vol. 125, no. Pt 16, pp. 3703-12, Aug 15 2012.
[84] D. A. De Ugarte et al., "Differential expression of stem cell mobilization-associated molecules on multi-lineage cells from adipose tissue and bone marrow," (in eng), Immunology letters, vol. 89, no. 2-3, pp. 267-70, Oct 31 2003.
[85] B. Annaz, K. A. Hing, M. Kayser, T. Buckland, and L. Di Silvio, "Porosity variation in hydroxyapatite and osteoblast morphology: a scanning electron microscopy study," (in eng), Journal of microscopy, vol. 215, no. Pt 1, pp. 100-10, Jul 2004.
[86] Y. Y. Shi, R. P. Nacamuli, A. Salim, and M. T. Longaker, "The osteogenic potential of adipose-derived mesenchymal cells is maintained with aging," (in eng), Plastic and reconstructive surgery, vol. 116, no. 6, pp. 1686-96, Nov 2005.
[87] G. I. Im, Y. W. Shin, and K. B. Lee, "Do adipose tissue-derived mesenchymal stem cells have the same osteogenic and chondrogenic potential as bone marrow-derived cells?," (in eng), Osteoarthritis and cartilage, vol. 13, no. 10, pp. 845-53, Oct 2005.
[88] A. Higuchi, Q. D. Ling, S. T. Hsu, and A. Umezawa, "Biomimetic Cell Culture Proteins as Extracellular Matrices for Stem Cell Differentiation," (in English), Chem. Rev., Review vol. 112, no. 8, pp. 4507-4540, Aug 2012.
[89] N. Chevallier et al., "Osteoblastic differentiation of human mesenchymal stem cells with platelet lysate," (in eng), Biomaterials, vol. 31, no. 2, pp. 270-8, Jan 2010.
[90] B. Trappmann et al., "Extracellular-matrix tethering regulates stem-cell fate," (in eng), Nature materials, vol. 11, no. 7, pp. 642-9, May 27 2012.
[91] C. L. Chou et al., "Micrometer scale guidance of mesenchymal stem cells to form structurally oriented cartilage extracellular matrix," (in eng), Tissue engineering. Part A, vol. 19, no. 9-10, pp. 1081-90, May 2013.
[92] C. Frantz, K. M. Stewart, and V. M. Weaver, "The extracellular matrix at a glance," (in eng), J Cell Sci, vol. 123, no. Pt 24, pp. 4195-200, Dec 15 2010.
[93] B. P. Chan and K. W. Leong, "Scaffolding in tissue engineering: general approaches and tissue-specific considerations," (in eng), European spine journal : official publication of the European Spine Society, the European Spinal Deformity Society, and the European Section of the Cervical Spine Research Society, vol. 17 Suppl 4, pp. 467-79, Dec 2008.
[94] C. W. Cheng, L. D. Solorio, and E. Alsberg, "Decellularized tissue and cell-derived extracellular matrices as scaffolds for orthopaedic tissue engineering," (in eng), Biotechnology advances, vol. 32, no. 2, pp. 462-84, Mar-Apr 2014.
[95] C. S. Hughes, L. M. Postovit, and G. A. Lajoie, "Matrigel: a complex protein mixture required for optimal growth of cell culture," (in eng), Proteomics, vol. 10, no. 9, pp. 1886-90, May 2010.
[96] F. Guilak, D. M. Cohen, B. T. Estes, J. M. Gimble, W. Liedtke, and C. S. Chen, "Control of stem cell fate by physical interactions with the extracellular matrix," (in eng), Cell Stem Cell, vol. 5, no. 1, pp. 17-26, Jul 2 2009.
[97] C. Gao, S. Peng, P. Feng, and C. Shuai, "Bone biomaterials and interactions with stem cells," (in eng), Bone research, vol. 5, p. 17059, 2017.
[98] B. N. Brown and S. F. Badylak, "Extracellular matrix as an inductive scaffold for functional tissue reconstruction," (in English), Transl. Res., Review vol. 163, no. 4, pp. 268-285, Apr 2014.
[99] F. Gattazzo, A. Urciuolo, and P. Bonaldo, "Extracellular matrix: a dynamic microenvironment for stem cell niche," (in eng), Biochimica et biophysica acta, vol. 1840, no. 8, pp. 2506-19, Aug 2014.
[100] M. D. Shoulders and R. T. Raines, "Collagen structure and stability," (in eng), Annual review of biochemistry, vol. 78, pp. 929-58, 2009.
[101] J. K. Mouw, G. Ou, and V. M. Weaver, "Extracellular matrix assembly: a multiscale deconstruction," (in eng), Nature reviews. Molecular cell biology, vol. 15, no. 12, pp. 771-85, Dec 2014.
[102] C. F. Marques, G. S. Diogo, S. Pina, J. M. Oliveira, T. H. Silva, and R. L. Reis, "Collagen-based bioinks for hard tissue engineering applications: a comprehensive review," (in eng), Journal of materials science. Materials in medicine, vol. 30, no. 3, p. 32, Mar 6 2019.
[103] T. H. Silva, J. Moreira-Silva, A. L. Marques, A. Domingues, Y. Bayon, and R. L. Reis, "Marine origin collagens and its potential applications," (in eng), Marine drugs, vol. 12, no. 12, pp. 5881-901, Dec 5 2014.
[104] R. E. Burgeson and M. E. Nimni, "Collagen types. Molecular structure and tissue distribution," (in eng), Clinical orthopaedics and related research, no. 282, pp. 250-72, Sep 1992.
[105] G. Vaissiere, B. Chevallay, D. Herbage, and O. Damour, "Comparative analysis of different collagen-based biomaterials as scaffolds for long-term culture of human fibroblasts," (in eng), Medical & biological engineering & computing, vol. 38, no. 2, pp. 205-10, Mar 2000.
[106] S. Chattopadhyay and R. T. Raines, "Review collagen-based biomaterials for wound healing," (in eng), Biopolymers, vol. 101, no. 8, pp. 821-33, Aug 2014.
[107] L. Cen, W. Liu, L. Cui, W. Zhang, and Y. Cao, "Collagen tissue engineering: development of novel biomaterials and applications," (in eng), Pediatric research, vol. 63, no. 5, pp. 492-6, May 2008.
[108] A. L. Fidler, S. P. Boudko, A. Rokas, and B. G. Hudson, "The triple helix of collagens - an ancient protein structure that enabled animal multicellularity and tissue evolution," (in eng), J Cell Sci, vol. 131, no. 7, Apr 9 2018.
[109] C. A. Sevilla, D. Dalecki, and D. C. Hocking, "Extracellular matrix fibronectin stimulates the self-assembly of microtissues on native collagen gels," (in eng), Tissue engineering. Part A, vol. 16, no. 12, pp. 3805-19, Dec 2010.
[110] P. Singh, C. Carraher, and J. E. Schwarzbauer, "Assembly of Fibronectin Extracellular Matrix," in Annual Review of Cell and Developmental Biology, Vol 26, vol. 26, R. Schekman, L. Goldstein, and R. Lehmann, Eds. (Annual Review of Cell and Developmental Biology. Palo Alto: Annual Reviews, 2010, pp. 397-419.
[111] E. Ruoslahti and M. D. Pierschbacher, "New perspectives in cell adhesion: RGD and integrins," (in eng), Science, vol. 238, no. 4826, pp. 491-7, Oct 23 1987.
[112] F. Li, S. D. Redick, H. P. Erickson, and V. T. Moy, "Force measurements of the alpha5beta1 integrin-fibronectin interaction," (in eng), Biophysical journal, vol. 84, no. 2 Pt 1, pp. 1252-62, Feb 2003.
[113] K. Somersalo and E. Saksela, "Fibronectin facilitates the migration of human natural killer cells," (in eng), European journal of immunology, vol. 21, no. 1, pp. 35-42, Jan 1991.
[114] R. S. Aziz-Seible and C. A. Casey, "Fibronectin: functional character and role in alcoholic liver disease," (in eng), World J Gastroenterol, vol. 17, no. 20, pp. 2482-99, May 28 2011.
[115] D. C. Hocking, J. Sottile, and K. J. Langenbach, "Stimulation of integrin-mediated cell contractility by fibronectin polymerization," (in eng), The Journal of biological chemistry, vol. 275, no. 14, pp. 10673-82, Apr 7 2000.
[116] K. T. Preissner, "The role of vitronectin as multifunctional regulator in the hemostatic and immune systems," (in eng), Blut, vol. 59, no. 5, pp. 419-31, Nov 1989.
[117] E. Spreghini, A. Gismondi, M. Piccoli, and G. Santoni, "Evidence for alphavbeta3 and alphavbeta5 integrin-like vitronectin (VN) receptors in Candida albicans and their involvement in yeast cell adhesion to VN," (in eng), The Journal of infectious diseases, vol. 180, no. 1, pp. 156-66, Jul 1999.
[118] H. J. Garrigues, Y. E. Rubinchikova, C. M. Dipersio, and T. M. Rose, "Integrin alphaVbeta3 Binds to the RGD motif of glycoprotein B of Kaposi′s sarcoma-associated herpesvirus and functions as an RGD-dependent entry receptor," (in eng), Journal of virology, vol. 82, no. 3, pp. 1570-80, Feb 2008.
[119] M. Rusnati, E. Tanghetti, P. Dell′Era, A. Gualandris, and M. Presta, "alphavbeta3 integrin mediates the cell-adhesive capacity and biological activity of basic fibroblast growth factor (FGF-2) in cultured endothelial cells," (in eng), Mol Biol Cell, vol. 8, no. 12, pp. 2449-61, Dec 1997.
[120] L. F. Reichardt and K. J. Tomaselli, "Extracellular matrix molecules and their receptors: functions in neural development," (in eng), Annual review of neuroscience, vol. 14, pp. 531-70, 1991.
[121] D. Jenne and K. K. Stanley, "Nucleotide sequence and organization of the human S-protein gene: repeating peptide motifs in the "pexin" family and a model for their evolution," (in eng), Biochemistry, vol. 26, no. 21, pp. 6735-42, Oct 20 1987.
[122] C. Selleri et al., "Involvement of the urokinase-type plasminogen activator receptor in hematopoietic stem cell mobilization," (in eng), Blood, vol. 105, no. 5, pp. 2198-205, Mar 1 2005.
[123] K. T. Preissner, "Structure and biological role of vitronectin," (in eng), Annual review of cell biology, vol. 7, pp. 275-310, 1991.
[124] C. D. Madsen, G. M. Ferraris, A. Andolfo, O. Cunningham, and N. Sidenius, "uPAR-induced cell adhesion and migration: vitronectin provides the key," (in eng), The Journal of cell biology, vol. 177, no. 5, pp. 927-39, Jun 4 2007.
[125] H. K. Makadia and S. J. Siegel, "Poly Lactic-co-Glycolic Acid (PLGA) as Biodegradable Controlled Drug Delivery Carrier," (in eng), Polymers, vol. 3, no. 3, pp. 1377-1397, Sep 1 2011.
[126] P. Gentile, V. Chiono, I. Carmagnola, and P. V. Hatton, "An overview of poly(lactic-co-glycolic) acid (PLGA)-based biomaterials for bone tissue engineering," (in eng), International journal of molecular sciences, vol. 15, no. 3, pp. 3640-59, Feb 28 2014.
[127] I. Bruzauskaite, D. Bironaite, E. Bagdonas, and E. Bernotiene, "Scaffolds and cells for tissue regeneration: different scaffold pore sizes-different cell effects," (in eng), Cytotechnology, vol. 68, no. 3, pp. 355-69, May 2016.
[128] S. Tiwari, R. Patil, and P. Bahadur, "Polysaccharide Based Scaffolds for Soft Tissue Engineering Applications," (in eng), Polymers, vol. 11, no. 1, Dec 20 2018.
[129] R. P. Felix Lanao, A. M. Jonker, J. G. Wolke, J. A. Jansen, J. C. van Hest, and S. C. Leeuwenburgh, "Physicochemical properties and applications of poly(lactic-co-glycolic acid) for use in bone regeneration," (in eng), Tissue engineering. Part B, Reviews, vol. 19, no. 4, pp. 380-90, Aug 2013.
[130] J. M. Lu et al., "Current advances in research and clinical applications of PLGA-based nanotechnology," (in eng), Expert review of molecular diagnostics, vol. 9, no. 4, pp. 325-41, May 2009.
[131] M. J. R. Virlan et al., "Current Uses of Poly(lactic-co-glycolic acid) in the Dental Field: A Comprehensive Review," (in English), J. Chem., Review p. 12, 2015, Art no. 525832.
[132] P. C. Baer and H. Geiger, "Adipose-derived mesenchymal stromal/stem cells: tissue localization, characterization, and heterogeneity," (in eng), Stem Cells Int, vol. 2012, p. 812693, 2012.
[133] C. M. Lo, H. B. Wang, M. Dembo, and Y. L. Wang, "Cell movement is guided by the rigidity of the substrate," (in eng), Biophysical journal, vol. 79, no. 1, pp. 144-52, Jul 2000
指導教授 樋口亞紺(Akon Higuchi) 審核日期 2019-8-20
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