博碩士論文 103324601 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:21 、訪客IP:3.142.43.151
姓名 沐督利(Saradaprasan Muduli)  查詢紙本館藏   畢業系所 化學工程與材料工程學系
論文名稱 人類羊水間葉幹細胞培養於具有奈米片段與最佳表面硬度的生醫材料,其增殖與成骨分化能力
(Proliferation and Osteogenic Differentiation of Human Amniotic Fluid derived Stem Cells Cultured on Biomaterials Having Nanosegments and Optimal Elasticity)
相關論文
★ 人類幹細胞培養於熱敏感奈米片段材料之研究★ 抗癌藥物的鑑定性用於分析大腸癌中癌幹細胞之研究
★ 利用具有奈米片段的生醫材料進行純化及去除癌症幹細胞★ 羊水間葉幹細胞培養於細胞外間質及材料硬度/彈性表面,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★ 多能幹細胞在無異種條件下分化為間充質幹細 胞的生物材料比較研究
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) 人類羊水間葉幹細胞(hAFSCs)為多能性胎兒細胞,具有能夠分化成特定細胞的能力,包括代表性的三個胚層。AFSCs可能成為幹細胞在再生醫學和組織工程更合適的來源。然而,由於幹細胞的特性,如適當的分化和多能性的維持,不只調節幹細胞本身,也可利用細胞的培養環境。此外,像是調整細胞培養基材的物理特性如軟硬度,可能會影響幹細胞分化的命運。在這個研究裡,調配在基材上不同的軟硬度,並固定有細胞外基質衍生的寡肽的細胞培養基材培養hAFSCs的成骨分化的效率。利用調配具有不同的軟硬度的polyvinylalcohol-co-itaconic acid(PVA-IA)在培養皿上,通過控制交聯劑的交聯時間製備。將PVA-IA培養皿通過N-(3-dimethylaminopropyl)-N’-ethylcarbodiimide hydrochloride ((EDC)和N-hydroxysuccinimide(NHS)去接枝上細胞外基質(ECM)衍生的寡肽。在這項研究中選擇了5種不同類型的寡肽,維持 AFSCs的多能性,藉由測定qRT-PCR測量,利用多能性基因的表現(Nanog的,Oct4和Sox2的)找出細胞培養基質的最佳軟硬度。為了分析hAFSCs的成骨分化特性,在兩週和四周利用誘導培養液誘導分化後進行alkaline phosphatase(ALP),alizarin Red S staining和von Kossa staining。利用物理特性如培養材料的軟硬度以及細胞外基質成分的生物特性可以誘導並決定hAFSCs分化成成骨細胞。
摘要(英) Human amniotic fluid-derived stem cells (hAFSCs) are pluripotent fetal cells capable of differentiation into multiple lineages, including representatives of the three embryonic germ layers. AFSCs may become a more suitable source of stem cells in regenerative medicine and tissue engineering. However, stem cell characteristics, such as proper differentiation and maintenance of pluripotency, are regulated not only by the stem cells themselves but also by their microenvironment. Furthermore, physical characteristics of cell culture substrates such as substrate elasticity may influence the fate of stem cell differentiation. I investigated the efficiency of osteogenic differentiation of hAFSCs cultured on cell culture substrates which have different elasticities and are immobilized with extracellular matrix-derived oligopeptides. The dishes coated with polyvinylalcohol-co-itaconic acid (PVA-IA) films having different elasticities were prepared by controlling the crosslinking time in crosslinking solution that contains glutaraldehyde. The PVA-IA dishes were grafted with extracellular matrix (ECM)-derived oligopeptides through N-(3-dimethylaminopropyl)-N’-ethylcarbodiimide hydrochloride (EDC) and N-hydroxysuccinimide (NHS) chemistry in an aqueous solution. Five different types of oligopeptides were selected in this study. Pluripotent gene expressions (Nanog, Oct4 and Sox2) were evaluated by the qRT-PCR measurements. There was an optimal elasticity of cell culture matrix to keep pluripotency of AFSCs for their culture. To characterize osteogenic differentiation of hAFSCs, alkaline phosphatase (ALP) activity, alizarin Red S staining and von Kossa staining were evaluated after two weeks and four weeks of culture in induction media. It is suggested that physical cues such as stiffness of culture materials as well as biological cues of extracellular matrix components can guide and decide differentiation of hAFSCs into osteoblasts.
關鍵字(中) ★ 人類羊水間葉幹細胞
★ 細胞外基質
關鍵字(英) ★ Human amniotic fluid derived stem cells
★ Extracellular matrix
論文目次 Chapter 1: Introduction 1
1-1 Regenerative medicine 1
1-1-1 Use of stem cells in regenerative medicine 1
1-2 What is a stem cell? 2
1-2-1 Self-renewal 2
1-2-2 Potency 3
1-2-3 Where do stem cells come from? 3
1-3 Types of stem cells 4
1-3-1 Embryonic stem cells (ESCs) 4
1-3-2 Induced pluripotent stem cells (iPSCs) 6
1-3-3 Adult stem cells 7
1-4 Amniotic fluid 9
1-4-1 Definition 9
1-4-2 Contents of amniotic fluid 9
1-4-3 Clinical significance of amniotic fluid 10
1-4-4 Isolation of amniotic fluid by amniocentesis 10
1-5 Amniotic fluid-derived stem cells 11
1-5-1 Cells present in amniotic fluid 11
1-5-1 Cells present in amniotic fluid 12
1-5-2 Isolation of amniotic fluid derived stem cells (AFSCs) 15
1-5-3 Characterization of amniotic fluid derived stem cells 15
1-6 Stem cell microenvironments 19
1-6-1 Cell-soluble factor interactions 19
1-6-2 Cell-cell interactions 20
1-6-3 Cell-biomaterial interactions 20
1-7 ECM and ECM-mimicking oligopeptides 22
1-7-1 Type and classification of artificial ECMs 24
1-7-2 ECM protein-derived peptides 26
1-7-3 The effect of extracellular matrix (ECM) on stem cells 27
1-8 Osteogenic differentiation 28
1-8-1 The process of bone development in situ 28
1-8-2 Developmental pathways for bone formation 29
1-8-3 The marker of osteogenic differentiation 33
1-9 Cardiomyocyte differentiation 34
1-9-1 Developmental pathways for Heart formation 34
1-9-2 Transcription factor programs in the myocardium 35
1-9-3 Meta-analysis of protocols for cardiomyocyte differentiation of MSCs 36
Chapter 2: Materials and methods 41
2-1 Materials 41
2-1-1 Cell culture medium 41
2-1-3 Serum 41
2-1-4 Antibiotics 41
2-1-5 Growth factor 42
2-1-6 PVA-IA film 42
2-1-7 ECM-derived peptides 42
2-1-9 RNA extraction 43
2-1-10 Reverse transcription (RT) 43
2-1-11 Quantitative PCR (qPCR) 43
2-1-12 qPCR Probes 43
2-2 Methods and Analysis 44
2-2-1 PVA-IA film preparation 44
2-2-2 Preparation of PVA-IA surfaces grafted with ECM-derived oligopeptides 45
2-2-5 Phosphate buffered saline (PBS) preparation 46
2-2-6 Preparation of FGF-2 (b-FGF) protein stock solution 46
2-2-7 Preparation of 5-Azacytidine stock solution 46
2-2-8 Preparation of cell culture medium 46
2-2-9 Cell cultivation 47
2-2-10 Counting of total cell number 48
2-2-11 Isolation of total RNA 49
2-2-12 Reverse Transcription of mRNA into cDNA 50
2-2-12 Quantitative real time polymerase chain reaction 51
2-2-13 Imunofluorescence assay 53
2-2-14 Alkaline phosphatase activity 54
2-2-15 Alizarin Red staining 54
2-2-15 von Kossa staining 55
2-2-16 Quantitative analysis of osteogenesis 55
Chapter 3: Results and Discussion 56
3-1 Isolation of hAFSC from human amniotic fluid on surface modified biomaterials 56
3-2 Proliferation of hAFSCs on PVAIA hydrogels grafted with different oligopeptides having different elasticities 61
3-3 Pluripotency of hAFSCs cultured on PVA-IA hydrogels grafted with and without oligopeptides, which have different elasticities 72
3-4 Osteogenic differentiation of hAFSCs cultured on PVA-IA hydrogels grafted with and without oligopeptides, which have different elasticities 77
3-5 Cardiomyogenic differentiation of hAFSCs in xeno-free condition 88
Chapter 4: Conclusion 93
References 95











Index of Figures
Figure1-1 Bone marrow transplant. 2
Figure1-2 Isolation and culture of ESCs from blastocysts. 5
Figure1-3 Generation of induced pluripotent stem cells (iPSCs) [31]. 6
Figure1-4 Morphology of MSCs derived from human amniotic fluid. 7
Figure1- 5 MSC differentiation 8
Figure1-6 Human fetus surrounded by amniotic fluid. 9
Figure1-7 Isolation of amniotic fluid by amniocentesis. 11
Figure1-9 Mature primary colonies of amniotic fluid cells after 14-16 days in culture: (a) F-type, (b) AF-type, (c) E-type [37]. 13
Figure1-10 Surface markers and pluripotent markers expression of hAFSCs [16]. 16
Figure1-11 ECM proteins guide mesenchymal stem cell fate [30]. 18
Figure1-12 The microenvironment of stem cells [30]. 19
Figure1- 13 Tissue elasticity and differentiation of native MSCs [75]. 22
Figure1-14 Osteogenic differentiation of MSCs. 29
Figure1-15 Runx2 is a mediator of molecular switches for bone development [150]. 32
Figure1-16 Temporal prolife of homeodomain proteins during the BMP2 induced 32
Figure1-17 Model of a molecular pathway for cardiac development [203]. 36
Figure1-18 Meta analysis of protocols for cardiomyocyte differentiation of MSCs. 38
Figure 2-1 Preparation of surface modified dishes with different elasticities and grafted with different oligopeptides. 45
Figure 2-2 Hemocytometer with 18 grids to count total cell number. 49
Fig 3-2 Growth curve of primary (passage 0) hAFSCs on ECM-coating and non-coating TCPS dishes in DMEM medium with 20% FBS. 59
Fig 3-3 Doubling time of primary (passage 0) hAFSCs cultured on ECM-coating and non-coating TCPS dishes in DMEM medium with 20% FBS. 59
Fig 3-4 Detachment of primary (passage 0) hAFSCs from ECM-coating and non-coating TCPS dishes by treating trypsin/EDTA for 5 minutes. The hAFSCs attachment on the Synthemax-coated dish is the strongest compared to that of other ECM-coating and non-coating TCPS dishes. 60
Figure 3-5 Morphology of hAFSCs cultured on PVA-IA hydrogels grafted with and without oligovitronectin (oligoVN) which have different elasticities (10.6, 11.1, 12.2 and 18.3 kPa), in DMEM with 20% FBS on day 3 of passage 4. 63
Figure 3-6 Morphology of hAFSCs cultured on PVA-IA hydrogels grafted with and without oligovitronectin (oligo-VN) which have different elasticities (25.3 and 30.4 kPa) and on TCPS (3700 kPa), in DMEM with 20% FBS on day 3 of passage 4. 64
Figure 3-7 Morphology of hAFSCs cultured on PVA-IA hydrogels grafted with and without different oligopeptides which have elasticity of 12.2 kPa, in DMEM with 20% FBS on day 3 of passage 4. 65
Figure 3-8 Morphology of hAFSCs cultured on PVA-IA hydrogels grafted with and without different oligopeptides which have elasticity of 18.3 kPa, in DMEM with 20% FBS on day 3 of passage 4. 66
Figure 3-9 Morphology of hAFSCs cultured on PVA-IA hydrogels grafted with and without different oligopeptides which have elasticity of 25.3 kPa, in DMEM with 20% FBS on day 3 of passage 4. 67
Figure 3-10 Morphology of hAFSCs cultured on PVA-IA hydrogels grafted with and without different oligopeptides which have elasticity of 30.4 kPa, in DMEM with 20% FBS on day 3 of passage 4. 68
Figure 3-11 Growth curve of hAFSCs cultured on TCPS dishes and PVA-IA hydrogels grafted with and without oligopeptides having elasticity of 12.2 kPa in DMEM with 20% FBS at passage 4. 69
Figure 3-13 Growth curve of hAFSCs cultured on PVA-IA hydrogels grafted with and without oligopeptides which have elasticity of 25.3 kPa, in DMEM with 20% FBS at passage 4. 70
Figure 3-14 Growth curve of hAFSCs cultured on PVA-IA hydrogels grafted with and without different oligopeptides which have elasticity of 30.4 kPa, in DMEM with 20% FBS at passage 4.. 70
Figure 3-15 Doubling time of hAFSCs cultured on TCPS, PVA-IA hydrogels grafted with and without oligopeptides which have different elasticities (12.2, 19.6, 25.3 and 30.4kPa), in DMEM with 20% FBS at passage 4. 71
Figure 3-16 SOX2 gene expression of hAFSCs on PVA-IA hydrogels grafted with and without oligopeptides, which have different elasticities (12.2, 19.6, 25.3, and 30.4kPa). 73
Figure 3-17 OCT4 gene expression of hAFSCs on PVA-IA hydrogels grafted with and without oligopeptides, which have different elasticity (12.2, 19.6, 25.3, and 30.4kPa). 75
Figure 3-18 Timeline and characterization for osteogenic differentiation of hAFSCs. 79
Figure 3-19 ALP activity of hAFSCs on PVA-IA hydrogels grafted with and without oligo-VN, which have elasticity of 10.6 kPa (2h) to 30.4 kPa (48h) after culturing in osteogenic induction medium for 14 days. 79
Figure 3-20 ALP activity of hAFSCs on PVA-IA hydrogels grafted with different oligopeptides, which have different elasticities after culturing in osteogenic induction media for 14 days. 80
Figure 3-21 Alizarin Red S staining of hAFSCs cultured on PVA-IA hydrogels grafted with and without oligopeptides, which have different elasticities after culturing in osteogenic induction medium for 28 days. 83
Figure 3-22 Percentage of Alizarin Red S staining cells analyzed by using Image J system (NIH). 83
Figure 3-23 von-Kossa staining (calcium phosphate deposition) of hAFSCs cultured on PVA-IA hydrogels grafted with and without oligo-peptides, which have different elasticities after culturing in osteogenic induction media for 28 days. 86
Figure 3-24 Percentage of von-Kossa staining cells analyzed by using image J system (NIH). 86
Figure 3-25 Differentiation protocol for chemically defined generation of cardiomyocytes from hAFSCs. 89
Figure 3-26 Morphology of hAFSCs cultured on TCPS dishes in growth medium (MesenCult-xenofree medium) for 5 days (day -5 to day 0) before differentiating into cardiomyocytes and then cultured in differentiation medium (CDM3) for 10 days (day 0 to day 10). 90
Figure 3-27 Immunostaining of sarcomeric proteins (α-SMA & cTnT) on hAFSCs at day 4, day 8, day 10 and day 12 of cardiomyocyte differentiation in CDM3 medium. 92
參考文獻 References
[1] New health facility aims to translate stem cell science into therapies | USC News
[2] Mason C., Dunnill, P., A brief definition of regenerative medicine. Regenerative Medicine, 2008. 3(1): p. 1–5.
[3] Confer, D.L., et al, Twenty years of unrelated donor hematopoietic cell transplantation for adult recipients facilitated by the National Marrow Donor Program. Biology of Blood and Marrow Transplantation, 2008. 14 (9 Supplement): p. 8–15.
[4] Malard, F., et al, New Insight for the Diagnosis of Gastrointestinal Acute Graft-versus-Host Disease. Mediators of Inflammation, 2014. Article ID 701013, 9 pages.
[5] Weissman, I.L., et al, Stem and progenitor cells: origins, phenotypes, lineage commitments, and transdifferentiations. Annual Review of Cell and Developmental Biology, 2001. 17: p. 387-403.
[6] Morrison, S.J., et al, Mechanisms of Stem Cell Self-Renewal. Annual Review of Cell and Developmental Biology, 2009. 25: p. 377-406.
[7] What are the unique properties of all stem cells? In Stem Cell Information [World Wide Web site]. Bethesda, MD: National Institutes of Health, U.S. Department of Health and Human Services, 2015.
[8] Mitalipov, S., et al, Totipotency, pluripotency and nuclear reprogramming. Advances in Biochemical Engineering & Biotechnology, 2009. 114: p. 185–99.
[9] Binder, Marc D.; Hirokawa, Nobutaka; Uwe Windhorst, eds. (2009). Encyclopedia of neuroscience. Berlin: Springer. ISBN 978-3540237358
[10] Tallone T., et al, Adult human adipose tissue contains several types of multipotent cells. Journal of Cardiovascular Translational Research, 2011. 4 (2): p. 200–10.
[11] Hans RS. The potential of stem cells: An inventory. In: Knoepffler N, Schipanski D, Sorgner SL, editors. Human biotechnology as Social Challenge. England: Ashgate Publishing, Ltd; 2007. p. 28.
[12] http://gowiki.tamu.edu/wiki/index.php/Category:GO:0061017_!_hepatoblast_differentiation
[13] Reya, T., et al., Stem cells, cancer, and cancer stem cells. Nature, 2001. 414(6859): p. 105-111.
[14] Fauza, D., Amniotic fluid and placental stem cells. Best Practice & Research in Clinical Obstetrics & Gynaecology, 2004. 18(6): p. 877-891.
[15] Kolambkar, Y.M., et al., Chondrogenic differentiation of amniotic fluid-derived stem cells. Journal of Molecular Histology, 2007. 38(5): p. 405-413.
[16] De Coppi, P., et al., Isolation of amniotic stem cell lines with potential for therapy. Nature Biotechnology, 2007. 25(1): p. 100-106.
[17] Zheng, Y.-B., et al., Characterization and hepatogenic differentiation of mesenchymal stem cells from human amniotic fluid and human bone marrow: A comparative study. Cell Biology International, 2008. 32(11): p. 1439-1448.
[18] Tsai, M.-S., et al., Functional network analysis of the transcriptomes of mesenchymal stem cells derived from amniotic fluid, amniotic membrane, cord blood, and bone marrow. Stem Cells, 2007. 25(10): p. 2511-2523.
[19] Kim, J., et al., Human amniotic fluid-derived stem cells have characteristics of multipotent stem cells. Cell Proliferation, 2007. 40(1): p. 75-90.
[20] Battula, V.L., et al., Human placenta and bone marrow derived MSC cultured in serum-free, b-FGF-containing medium express cell surface frizzled-9 and SSEA-4 and give rise to multilinelage differentiation. Differentiation, 2007. 75(4): p. 279-291.
[21] Tsai, M.S., et al., Clonal amniotic fluid-derived stem cells express characteristics of both mesenchymal and neural stem cells. Biology of Reproduction, 2006. 74(3): p. 545-551.
[22] Poloni, A., et al., Characterization and expansion of mesenchymal progenitor cells from first-trimester chorionic villi of human placenta. Cytotherapy, 2008. 10(7): p. 690-697.
[23] Gucciardo, L., et al., Fetal mesenchymal stem cells: isolation, properties and potential use in perinatology and regenerative medicine. Bjog-an International Journal of Obstetrics and Gynaecology, 2009. 116(2): p. 166-172
[24] Klimanskaya, I., et al., Human embryonic stem cells derived without feeder cells. Lancet, 2005. 365(9471): p. 1636-1641.
[25] Higuchi, A., et al., Biomaterials for the Feeder-Free Culture of Human Embryonic Stem Cells and Induced Pluripotent Stem Cells. Chemical Reviews, 2011. 111(5): p. 3021-3035.
[26] Okita, K., T. Ichisaka, and S. Yamanaka, Generation of germline-competent induced pluripotent stem cells. Nature, 2007. 448(7151): p. 313-7.
[27] 20. Takahashi, K. and S. Yamanaka, Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell, 2006. 126(4): p. 663-676.
[28] Lin, S.-L., et al., Mir-302 reprograms human skin cancer cells into a pluripotent ES-cell-like state. Rna-a Publication of the Rna Society, 2008. 14(10): p. 2115-2124.
[29] Zhou, H., et al., Generation of Induced Pluripotent Stem Cells Using Recombinant Proteins (vol 4, pg 381, 2009). Cell Stem Cell, 2009. 4(6): p. 581-581
[30] Higuchi, A., et al., Biomimetic Cell Culture Proteins as Extracellular Matrices for Stem Cell Differentiation. Chemical Reviews, 2012. 112(8): p. 4507-4540.
[31] Meissner, A., et al, Induced pluripotent stem cells: current progress and potential for regenerative medicine. Trends in Molecular Medicine, 2009. 15(2): p. 59–68
[32] What are adult stem cells? In Stem Cell Information [World Wide Web site]. Bethesda, MD: National Institutes of Health, U.S. Department of Health and Human Services, 2015.
[33] Brighton, C.T. and R.M. Hunt, EARLY HISTOLOGICAL AND ULTRASTRUCTURAL-CHANGES IN MEDULLARY FRACTURE CALLUS. Journal of Bone and Joint Surgery-American Volume, 1991. 73A (6): p. 832-847.
[34] Chamberlain, G., et al., Concise review: Mesenchymal stem cells: Their phenotype, differentiation capacity, immunological features, and potential for homing. Stem Cells, 2007. 25(11): p. 2739-2749.
[35] Modena, A.B., Amniotic fluid dynamics. Acta Biomedica, 2004. 75(suppl): p. 11–13.
[36] Beall, M.H., Amniotic fluid dynamics. Placenta, 2007. 28: p. 816–823.
[37] Gosden, C.M., Amniotic fluid cell types and culture. British Medical Bulletin, 1983. 39(4): p. 348-354.
[38] Hoehn, H. and D. Salk, Morphological and biochemical heterogeneity of amniotic fluid cells in culture, in Methods in Cell Biology, A.L. Samuel and J.D. Gretchen, Editors. 1982. p. 11-34.
[39] http://www.health.harvard.edu/diagnostic-tests/amniosentesis.htm
[40] Ronnee K. Yashon; Michael R. Cummings (23 September 2011). Human Genetics and Society. Cengage Learning. p. 83. ISBN 978-0-538-73321-2.
[41] Tsai, M.S., et al., Isolation of human multipotent mesenchymal stem cells from second-trimester amniotic fluid using a novel two-stage culture protocol. Human Reproduction, 2004. 19(6): p. 1450-1456.
[42] Jiang, Y.H., et al., Pluripotency of mesenchymal stem cells derived from adult marrow. Nature, 2002. 418(6893): p. 41-49.
[43] Perrone, L., et al., Postnatal weight change is influenced by mother-newborn pair leptin levels. Nutrition Research, 2000. 20(11): p. 1531-1536.
[44] Zambotti, F., et al., Monoamine metabolites and related compounds in human amniotic fluid: Assay by gas chromatography and gas chromatography-mass spectrometry. Clinica Chimica Acta 1975. 61(3): p. 247-256.
[45] Barry, F.P., et al., The monoclonal antibody SH-2, raised against human mesenchymal stem cells, recognizes an epitope on endoglin (CD105). Biochemical and Biophysical Research Communications, 1999. 265(1): p. 134-139.
[46] Barry, F., et al., The SH-3 and SH-4 antibodies recognize distinct epitopes on CD73 from human mesenchymal stem cells. Biochemical and Biophysical Research Communications, 2001. 289(2): p. 519-524.
[47] Baksh, D., L. Song, and R.S. Tuan, Adult mesenchymal stem cells: characterization, differentiation, and application in cell and gene therapy. Journal of Cellular and Molecular Medicine, 2004. 8(3): p. 301-316.
[48] Ririe, K.M., R.P. Rasmussen, and C.T. Wittwer, Product differentiation by analysis of DNA melting curves during the polymerase chain reaction. Analytical Biochemistry, 1997. 245(2): p. 154-160.
[49] Shamblott, M.J., et al., Human embryonic germ cell derivatives express a broad range of developmentally distinct markers and proliferate extensively in vitro. Proceedings of the National Academy of Sciences of the United States of America, 2001. 98(1): p. 113-118.
[50] Pittenger, M.F., et al., Multilineage potential of adult human mesenchymal stem cells. Science, 1999. 284(5411): p. 143-147.
[51] Zuk, P.A., et al., Human adipose tissue is a source of multipotent stem cells. Molecular Biology of the Cell, 2002. 13(12): p. 4279-4295.
[52] Engler, A.J., et al., Matrix elasticity directs stem cell lineage specification. Cell, 2006. 126(4): p. 677-689.
[53] Higuchi, A., et al., Physical Cues of Biomaterials Guide Stem Cell Differentiation Fate. Chemical Reviews, 2013. 113(5): p. 3297-3328.
[54] Salasznyk, R.M., et al., Adhesion to vitronectin and collagen I promotes osteogenic differentiation of human mesenchymal stem cells. Journal of Biomedicine and Biotechnology, 2004(1): p. 24-34.
[55] Hashimoto, J., Y. Kariya, and K. Miyazaki, Regulation of proliferation and chondrogenic differentiation of human mesenchymal stem cells by laminin-5 (laminin-332). Stem Cells, 2006. 24(11): p. 2346-2354.
[56] Chastain, S.R., et al., Adhesion of mesenchymal stem cells to polymer scaffolds occurs via distinct ECM ligands and controls their osteogenic differentiation. Journal of Biomedical Materials Research Part A, 2006. 78A(1): p. 73-85.
[57] Suzuki, S., et al., Effects of Extracellular Matrix on Differentiation of Human Bone Marrow-Derived Mesenchymal Stem Cells into Smooth Muscle Cell Lineage: Utility for Cardiovascular Tissue Engineering. Cells Tissues Organs, 2010. 191(4): p. 269-280.
[58] van Dijk, A., et al., Differentiation of human adipose-derived stem cells towards cardiomyocytes is facilitated by laminin. Cell and Tissue Research, 2008. 334(3): p. 457-467.
[59] Mruthyunjaya, S., et al., Laminin-1 induces neurite outgrowth in human mesenchymal stem cells in serum/differentiation factors-free conditions through activation of FAK-MEK/ERK signaling pathways. Biochemical and Biophysical Research Communications, 2010. 391(1): p. 43-48.
[60] Delcroix, G.J.R., et al., The therapeutic potential of human multipotent mesenchymal stromal cells combined with pharmacologically active microcarriers transplanted in hemi-parkinsonian rats. Biomaterials, 2011. 32(6): p. 1560-1573.
[61] Levenberg, S., et al., Differentiation of human embryonic stem cells on three-dimensional polymer scaffolds. Proceedings of the National Academy of Sciences of the United States of America, 2003. 100(22): p. 12741-12746.
[62] Park, I.-S., et al., The correlation between human adipose-derived stem cells differentiation and cell adhesion mechanism. Biomaterials, 2009. 30(36): p. 6835-6843.
[63] Ter Brugge, P.J. and J.A. Jansen, In vitro osteogenic differentiation of rat bone marrow cells subcultured with and without dexamethasone. Tissue Engineering, 2002. 8(2): p. 321-331.
[64] Park, J.S., et al., The effect of matrix stiffness on the differentiation of mesenchymal stem cells in response to TGF-beta. Biomaterials, 2011. 32(16): p. 3921-3930.
[65] Bhandari, D.R., et al., The simplest method for in vitro beta-cell production from human adult stem cells. Differentiation, 2011. 82(3): p. 144-152.
[66] Xie, Q.-P., et al., Human bone marrow mesenchymal stem cells differentiate into insulin-producing cells upon microenvironmental manipulation in vitro. Differentiation, 2009. 77(5): p. 483-491.
[67] Gabr, M.M., et al., Generation of Insulin-Producing Cells from Human Bone Marrow-Derived Mesenchymal Stem Cells: Comparison of Three Differentiation Protocols. Biomed Research International, 2014.
[68] Czubak, P., et al., A Modified Method of Insulin Producing Cells′ Generation from Bone Marrow-Derived Mesenchymal Stem Cells. Journal of Diabetes Research, 2014.
[69] Cooper, G.M., The Cell: A Molecular Approach. 2000-2013.
[70] Gilbert, P.M., et al., Substrate Elasticity Regulates Skeletal Muscle Stem Cell Self-Renewal in Culture. Science, 2010. 329(5995): p. 1078-1081.
[71] Georges, P.C., et al., Matrices with compliance comparable to that of brain tissue select neuronal over glial growth in mixed cortical cultures. Biophysical Journal, 2006. 90(8): p. 3012-3018.
[72] Flanagan, L.A., et al., Neurite branching on deformable substrates. Neuroreport, 2002. 13(18): p. 2411-2415.
[73] Hofstetter, C.P., et al., Marrow stromal cells form guiding strands in the injured spinal cord and promote recovery. Proceedings of the National Academy of Sciences of the United States of America, 2002. 99(4): p. 2199-2204.
[74] Kondo, T., et al., Sonic hedgehog and retinoic acid synergistically promote sensory fate specification from bone marrow-derived pluripotent stem cells. Proceedings of the National Academy of Sciences of the United States of America, 2005. 102(13): p. 4789-4794.
[75] Engler, A.J., et al., Myotubes differentiate optimally on substrates with tissue-like stiffness: pathological implications for soft or stiff microenvironments. Journal of Cell Biology, 2004. 166(6): p. 877-887.
[76] Ferrari, G., et al., Muscle regeneration by bone marrow derived myogenic progenitors. Science, 1998. 279(5356): p. 1528-1530.
[77] Andrades, J.A., et al., Selection and amplification of a bone marrow cell population and its induction to the chondro-osteogenic lineage by rhOP-1: an in vitro and in vivo study. International Journal of Developmental Biology, 2001. 45(4): p. 689-693.
[78] Holmbeck, K., et al., MT1-MMP-deficient mice develop dwarfism, osteopenia, arthritis, and connective tissue disease due to inadequate collagen turnover. Cell, 1999. 99(1): p. 81-92.
[79] Morinobu, M., et al., Osteopontin expression in osteoblasts and osteocytes during bone formation under mechanical stress in the calvarial suture in vivo. Journal of Bone and Mineral Research, 2003. 18(9): p. 1706-1715.
[80] Deng, J., et al., Mesenchymal stem cells spontaneously express neural proteins in culture and are neurogenic after transplantation. Stem Cells, 2006. 24(4): p. 1054-1064.
[81] McBeath, R., et al., Cell shape, cytoskeletal tension, and RhoA regulate stem cell lineage commitment. Developmental Cell, 2004. 6(4): p. 483-495.
[82] LaIuppa, J.A., et al., Culture materials affect ex vivo expansion of hematopoietic progenitor cells. Journal of Biomedical Materials Research, 1997. 36(3): p. 347-359.
[83] Seraj, M.J., et al., Functional evidence for a novel human breast carcinoma metastasis suppressor, BRMS1, encoded at chromosome 11q13. Cancer Research, 2000. 60(11): p. 2764-2769.
[84] Lu, Z., et al., Collagen Type II Enhances Chondrogenesis in Adipose Tissue-Derived Stem Cells by Affecting Cell Shape. Tissue Engineering Part A, 2010. 16(1): p. 81-90.
[85] Kikkawa, Y., et al., Integrin binding specificity of laminin-10/11 : laminin-10/11 are recognized by alpha 3 beta 1, alpha 6 beta 1 and alpha 6 beta 4 integrins. Journal of Cell Science, 2000. 113(5): p. 869-876.
[86] Jiang, X.S., et al., Surface-immobilization of adhesion peptides on substrate for ex vivo expansion of cryopreserved umbilical cord blood CD34(+) cells. Biomaterials, 2006. 27(13): p. 2723-2732.
[87] Gelain, F., et al., Designer Self-Assembling Peptide Nanofiber Scaffolds for Adult Mouse Neural Stem Cell 3-Dimensional Cultures. Plos One, 2006. 1(2).
[88] Frontiers in tissue engineering. 1998.
[89] Rosso, F., et al., From cell-ECM interactions to tissue engineering. Journal of Cellular Physiology, 2004. 199(2): p. 174-180.
[90] Langer, R. and J.P. Vacanti, Tissue engineering. Science, 1993. 260(5110): p. 920-926.
[91] Barbucci, R., Integrated Biomaterials Science. 2002.
[92] Putnam, A.J. and D.J. Mooney, Tissue engineering using synthetic extracellular matrices. Nature Medicine, 1996. 2(7): p. 824-826.
[93] Daley, W.P., S.B. Peters, and M. Larsen, Extracellular matrix dynamics in development and regenerative medicine. Journal of Cell Science, 2008. 121(3): p. 255-264.
[94] Rozario, T. and D.W. DeSimone, The extracellular matrix in development and morphogenesis: A dynamic view. Developmental Biology, 2010. 341(1): p. 126-140.
[95] Chen, L.-Y., et al., Effect of the surface density of nanosegments immobilized on culture dishes on ex vivo expansion of hematopoietic stem and progenitor cells from umbilical cord blood. Acta Biomaterialia, 2012. 8(5): p. 1749-1758.
[96] Koivunen, E., B.C. Wang, and E. Ruoslahti, Phage Libraries Displaying Cyclic Peptides with Different Ring Sizes: Ligand Specificities of the RGD-Directed Integrins. Bio-Technology, 1995. 13(3): p. 265-270.
[97] Kolhar, P., et al., Synthetic surfaces for human embryonic stem cell culture. Journal of Biotechnology, 2010. 146(3): p. 143-146.
[98] Klim, J.R., et al., A defined glycosaminoglycan-binding substratum for human pluripotent stem cells. Nature Methods, 2010. 7(12): p. 989-994.
[99] Rosso, F., et al., Smart materials as scaffolds for tissue engineering. Journal of Cellular Physiology, 2005. 203(3): p. 465-470.
[100] Moroni, L., J.R. De Wijn, and C.A. Van Blitterswijk, Integrating novel technologies to fabricate smart scaffolds. Journal of Biomaterials Science-Polymer Edition, 2008. 19(5): p. 543-572.
[101] Mano, J.F., et al., Natural origin biodegradable systems in tissue engineering and regenerative medicine: present status and some moving trends. Journal of the Royal Society Interface, 2007. 4(17): p. 999-1030.
[102] Lee, C.H., A. Singla, and Y. Lee, Biomedical applications of collagen. International Journal of Pharmaceutics, 2001. 221(1-2): p. 1-22.
[103] Pankov, R. and K.M. Yamada, Fibronectin at a glance. Journal of Cell Science, 2002. 115(20): p. 3861-3863.
[104] Mao, Y. and J.E. Schwarzbauer, Fibronectin fibrillogenesis, a cell-mediated matrix assembly process. Matrix Biology, 2005. 24(6): p. 389-399.
[105] http://www.sigmaaldrich.com/catalog/product/sigma/V8379?lang=en®ion=TW.
[106] Ogawa, T., et al., The short arm of laminin gamma 2 chain of laminin-5 (laminin-332) binds syndecan-1 and regulates cellular adhesion and migration by suppressing phosphorylation of integrin beta 4 chain. Molecular Biology of the Cell, 2007. 18(5): p. 1621-1633.
[107] Klees, R.F., et al., Laminin-5 induces osteogenic gene expression in human mesenchymal stem cells through an ERK-dependent pathway. Molecular Biology of the Cell, 2005. 16(2): p. 881-890.
[108] Ma, W., et al., Cell-extracellular matrix interactions regulate neural differentiation of human embryonic stem cells. Bmc Developmental Biology, 2008. 8.
[109] Tate, C.C., et al., Laminin and fibronectin scaffolds enhance neural stem cell transplantation into the injured brain. Journal of Tissue Engineering and Regenerative Medicine, 2009. 3(3): p. 208-217.
[110] Hayman, M.W., et al., Growth of human stem cell-derived neurons on solid three-dimensional polymers. Journal of Biochemical and Biophysical Methods, 2005. 62(3): p. 231-240.
[111] Martinez-Ramos, C., et al., Differentiation of postnatal neural stem cells into glia and functional neurons on laminin-coated polymeric substrates. Tissue Engineering Part A, 2008. 14(8): p. 1365-1375.
[112] Li, S., et al., Matrix assembly, regulation, and survival functions of laminin and its receptors in embryonic stem cell differentiation. Journal of Cell Biology, 2002. 157(7): p. 1279-1290.
[113] Hall, P.E., et al., Laminin enhances the growth of human neural stem cells in defined culture media. Bmc Neuroscience, 2008. 9.
[114] Delcroix, G.J.R., et al., Adult cell therapy for brain neuronal damages and the role of tissue engineering. Biomaterials, 2010. 31(8): p. 2105-2120.
[115] Kleinman, H.K. and G.R. Martin, Matrigel: Basement membrane matrix with biological activity. Seminars in Cancer Biology, 2005. 15(5): p. 378-386.
[116] Haque, A., et al., The effect of recombinant E-cadherin substratum on the differentiation of endoderm-derived hepatocyte-like cells from embryonic stem cells. Biomaterials, 2011. 32(8): p. 2032-2042.
[117] Kaur, G., et al., The promotion of osteoblastic differentiation of rat bone marrow stromal cells by a polyvalent plant mosaic virus. Biomaterials, 2008. 29(30): p. 4074-4081.
[118] Yue, X.-S., et al., A fusion protein N-cadherin-Fc as an artificial extracellular matrix surface for maintenance of stem cell features. Biomaterials, 2010. 31(20): p. 5287-5296.
[119] Shi, C., et al., Stem-cell-capturing collagen scaffold promotes cardiac tissue regeneration. Biomaterials, 2011. 32(10): p. 2508-2515.
[120] Lee, H.J., et al., Enhanced Chondrogenesis of Mesenchymal Stem Cells in Collagen Mimetic Peptide-Mediated Microenvironment. Tissue Engineering Part A, 2008. 14(11): p. 1843-1851.
[121] You, M., et al., Chondrogenic differentiation of human bone marrow mesenchymal stem cells on polyhydroxyalkanoate (PHA) scaffolds coated with PHA granule binding protein PhaP fused with RGD peptide. Biomaterials, 2011. 32(9): p. 2305-2313.
[122] Hennessy, K.M., et al., The effect of collagen I mimetic peptides on mesenchymal stem cell adhesion and differentiation, and on bone formation at hydroxyapatite surfaces. Biomaterials, 2009. 30(10): p. 1898-1909.
[123] Yang, F., et al., The effect of incorporating RGD adhesive peptide in polyethylene glycol diacrylate hydrogel on osteogenesis of bone marrow stromal cells. Biomaterials, 2005. 26(30): p. 5991-5998.
[124] Nguyen, L.H., et al., Unique biomaterial compositions direct bone marrow stem cells into specific chondrocytic phenotypes corresponding to the various zones of articular cartilage. Biomaterials, 2011. 32(5): p. 1327-1338.
[125] Betre, H., et al., Chondrocytic differentiation of human adipose-derived adult stem cells in elastin-like polypeptide. Biomaterials, 2006. 27(1): p. 91-99.
[126] Meinel, L., et al., Engineering bone-like tissue in vitro using human bone marrow stem cells and silk scaffolds. Journal of Biomedical Materials Research Part A, 2004. 71A(1): p. 25-34.
[127] Santiago, L.Y., et al., Peptide-surface modification of poly(caprolactone) with laminin-derived sequences for adipose-derived stem cell applications. Biomaterials, 2006. 27(15): p. 2962-2969.
[128] Wojtowicz, A.M., et al., Coating of biomaterial scaffolds with the collagen-mimetic peptide GFOGER for bone defect repair. Biomaterials, 2010. 31(9): p. 2574-2582.
[129] Cooke, M.J., et al., Neural differentiation regulated by biomimetic surfaces presenting motifs of extracellular matrix proteins. Journal of Biomedical Materials Research Part A, 2010. 93A(3): p. 824-832.
[130] Anderson, J.M., et al., Osteogenic Differentiation of Human Mesenchymal Stem Cells Directed by Extracellular Matrix-Mimicking Ligands in a Biomimetic Self-Assembled Peptide Amphiphile Nanomatrix. Biomacromolecules, 2009. 10(10): p. 2935-2944.
[131] Bhatnagar, R.S., J.J. Qian, and C.A. Gough, The role in cell binding of a beta-bend within the triple helical region in collagen alpha 1(I) chain: Structural and biological evidence for conformational tautomerism on fiber surface. Journal of Biomolecular Structure & Dynamics, 1997. 14(5): p. 547-60.
[132] Higuchi, A., et al., Polymeric Materials for Ex vivo Expansion of Hematopoietic Progenitor and Stem Cells. Polymer Reviews, 2009. 49(3): p. 181-200.
[133] Melkoumian, Z., et al., Synthetic peptide-acrylate surfaces for long-term self-renewal and cardiomyocyte differentiation of human embryonic stem cells. Nature Biotechnology, 2010. 28(6): p. 606-610.
[134] Pierschbacher, M.D. and E. Ruoslahti, Cell attachment activity of fibronectin can be duplicated by small synthetic fragments of the molecule. Nature, 1984. 309(5963): p. 30-33.
[135] Suzuki, S., et al., Complete amino acid sequence of human vitronectin deduced from cDNA. Similarity of cell attachment sites in vitronectin and fibronectin. The Embo Journal, 1985. 4(10): p. 2519-2524.
[136] Oldberg, A., A. Franzén, and D. Heinegård, The primary structure of a cell-binding bone sialoprotein. The Journal of Biological Chemistry, 1988. 263(36): p. 19430-19432.
[137] Scadden, D.T., The stem-cell niche as an entity of action. Nature, 2006. 441(7097): p. 1075-1079.
[138] Nilsson, S.K., et al., Osteopontin, a key component of the hematopoietic stem cell niche and regulator of primitive hematopoietic progenitor cells. Blood, 2005. 106(4): p. 1232-1239.
[139] Moore, K.A. and I.R. Lemischka, Stem cells and their niches. Science, 2006. 311(5769): p. 1880-1885.
[140] Schofield, R., The relationship between the spleen colony-forming cell and the haemopoietic stem cell. Blood Cells, 1978. 4(1-2): p. 7-25.
[141] Li, L.H. and T. Xie, Stem cell niche: Structure and function, in Annual Review of Cell and Developmental Biology. 2005. p. 605-631.
[142] Jensen, U.B., S. Lowell, and F.M. Watt, The spatial relationship between stem cells and their progeny in the basal layer of human epidermis: a new view based on whole-mount labelling and lineage analysis. Development, 1999. 126(11): p. 2409-2418.
[143] Nuttelman, C.R., M.C. Tripodi, and K.S. Anseth, Synthetic hydrogel niches that promote hMSC viability. Matrix Biology, 2005. 24(3): p. 208-218.
[144] Feng, Q., et al., Expansion of engrafting human hematopoietic stem/progenitor cells in three-dimensional scaffolds with surface-immobilized fibronectin. Journal of Biomedical Materials Research Part A, 2006. 78A(4): p. 781-791.
[145] Gerecht, S., et al., Hyaluronic acid hydrogen for controlled self-renewal and differentiation of human embryonic stem cells. Proceedings of the National Academy of Sciences of the United States of America, 2007. 104(27): p. 11298-11303.
[146] Chang, C.-F., et al., Three-dimensional collagen fiber remodeling by mesenchymal stem cells requires the integrin-matrix interaction. Journal of Biomedical Materials Research Part A, 2007. 80A(2): p. 466-474.
[147] Bellamkonda, R., et al., Laminin Oligopeptide Derivatized Agarose Gels Allow Three-Dimensional Neurite Extension In Vitro. Journal of Neuroscience Research, 1995. 41: p. 501-509.
[148] Olsen, B.R., A.M. Reginato, and W.F. Wang, Bone development. Annual Review of Cell and Developmental Biology, 2000. 16: p. 191-220.
[149] Harada, S. and G.A. Rodan, Control of osteoblast function and regulation of bone mass. Nature, 2003. 423(6937): p. 349-355.
[150] Lian, J.B., et al., Networks and hubs for the transcriptional control of osteoblastogenesis. Reviews in Endocrine & Metabolic Disorders, 2006. 7(1-2): p. 1-16.
[151] Ducy, P., et al., Osf2/Cbfa1: A transcriptional activator of osteoblast differentiation. Cell, 1997. 89(5): p. 747-754.
[152] Lengner, C.J., et al., Activation of the bone-related Runx2/Cbfa1 promoter in mesenchymal condensations and developing chondrocytes of the axial skeleton. Mechanisms of Development, 2002. 114(1-2): p. 167-170.
[153] Romero-Prado, M., et al., Functional characterization of human mesenchymal stem cells that maintain osteochondral fates. Journal of Cellular Biochemistry, 2006. 98(6): p. 1457-1470.
[154] Murtaugh, L.C., et al., The chick transcriptional repressor Nkx3.2 acts downstream of Shh to promote BMP-Dependent axial chondrogenesis. Developmental Cell, 2001. 1(3): p. 411-422.
[155] Iwamoto, M., et al., Runx2 expression and action in chondrocytes are regulated by retinoid signaling and parathyroid hormone-related peptide (PTHrP). Osteoarthritis and Cartilage, 2003. 11(1): p. 6-15.
[156] Eames, B.F., P.T. Sharpe, and J.A. Helms, Hierarchy revealed in the specification of three skeletal fates by Sox9 and Runx2. Developmental Biology, 2004. 274(1): p. 188-200.
[157] Lengner, C.J., et al., Nkx3.2-mediated repression of Runx2 promotes chondrogenic differentiation. Journal of Biological Chemistry, 2005. 280(16): p. 15872-15879.
[158] Guo, J., et al., PTH/PTHrP receptor delays chondrocyte hypertrophy via both Runx2-dependent and -independent pathways. Developmental Biology, 2006. 292(1): p. 116-128.
[159] Provot, S., et al., Nkx3.2/Bapx1 acts as a negative regulator of chondrocyte maturation. Development, 2006. 133(4): p. 651-662.
[160] Li, T.F., et al., Parathyroid hormone-related peptide (PTHrP) inhibits Runx2 expression through the PKA signaling pathway. Experimental Cell Research, 2004. 299(1): p. 128-136.
[161] Zelzer, E., et al., Tissue specific regulation of VEGF expression during bone development requires Cbfa1/Runx2. Mechanisms of Development, 2001. 106(1-2): p. 97-106.
[162] Pratap, J., et al., The Runx2 osteogenic transcription factor regulates matrix metalloproteinase 9 in bone metastatic cancer cells and controls cell invasion. Molecular and Cellular Biology, 2005. 25(19): p. 8581-8591.
[163] ten Dijke, P., et al., Signal transduction of bone morphogenetic proteins in osteoblast differentiation. Journal of Bone and Joint Surgery-American Volume, 2003. 85A: p. 34-38.
[164] Chen, D., M. Zhao, and G.R. Mundy, Bone morphogenetic proteins. Growth Factors, 2004. 22(4): p. 233-241.
[165] Bidder, M., T. Latifi, and D.A. Towler, Reciprocal temporospatial patterns of Msx2 and osteocalcin gene expression during murine odontogenesis. Journal of Bone and Mineral Research, 1998. 13(4): p. 609-619.
[166] Ferrari, D., et al., Dlx-5 in limb initiation in the chick embryo. Developmental Dynamics, 1999. 216(1): p. 10-15.
[167] Holleville, N., et al., BMP signals regulate Dlx5 during early avian skull development. Developmental Biology, 2003. 257(1): p. 177-189.
[168] Hassan, M.Q., et al., Dlx3 transcriptional regulation of osteoblast differentiation: Temporal recruitment of Msx2 Dlx3 an Dlx5 homeodomain proteins to chromatin of the osteocalcin gene. Molecular and Cellular Biology, 2004. 24(20): p. 9248-9261.
[169] Depew, M.J., et al., Reassessing the Dlx code: the genetic regulation of branchial arch skeletal pattern and development. Journal of Anatomy, 2005. 207(5): p. 501-561.
[170] Dodig, M., et al., Ectopic Msx2 overexpression inhibits and Msx2 antisense stimulates calvarial osteoblast differentiation. Developmental Biology, 1999. 209(2): p. 298-307.
[171] Lee, M.H., et al., BMP-2-induced Runx2 expression is mediated by Dlx5, and TGF-beta 1 opposes the BMP-2-induced osteoblast differentiation by suppression of Dlx5 expression. Journal of Biological Chemistry, 2003. 278(36): p. 34387-34394.
[172] Lee, M.H., et al., BMP-2-induced Osterix expression is mediated by Dlx5 but is independent of Runx2. Biochemical and Biophysical Research Communications, 2003. 309(3): p. 689-694.
[173] Cheng, S.L., et al., Msx2 promotes osteogenesis and suppresses adipogenic differentiation of multipotent mesenchymal progenitors. Journal of Biological Chemistry, 2003. 278(46): p. 45969-45977.
[174] Ichida, F., et al., Reciprocal roles of Msx2 in regulation of osteoblast and adipocyte differentiation. Journal of Biological Chemistry, 2004. 279(32): p. 34015-34022.
[175] Yoshizawa, T., et al., Homeobox protein Msx2 acts as a molecular defense mechanism for preventing ossification in ligament fibroblasts. Molecular and Cellular Biology, 2004. 24(8): p. 3460-3472.
[176] Balint, E., et al., Phenotype discovery by gene expression profiling: Mapping of biological processes linked to BMP-2-mediated osteoblast differentiation. Journal of Cellular Biochemistry, 2003. 89(2): p. 401-426.
[177] Shirakabe, K., et al., Regulation of the activity of the transcription factor Runx2 by two homeobox proteins, Msx2 and Dlx5. Genes to Cells, 2001. 6(10): p. 851-856.
[178] Hassan, M.Q.e.a., hoxa10: a BMP-2-responsive gene activates runx2 and regulates osteogenesis. Journal of Bone and Mineral Research, 2005. 20(9): p. S5-S5.
[179] Lee, M.H., et al., Dlx5 specifically regulates Runx2 type II expression by binding to homeodomain-response elements in the Runx2 distal promoter. Journal of Biological Chemistry, 2005. 280(42): p. 35579-35587.
[180] Nakashima, K., et al., The novel zinc finger-containing transcription factor Osterix is required for osteoblast differentiation and bone formation. Cell, 2002. 108(1): p. 17-29.
[181] Pratap, J., et al., Cell growth regulatory role of Runx2 during proliferative expansion of preosteoblasts. Cancer Research, 2003. 63(17): p. 5357-5362.
[182] Kim, Y.J., et al., The bone-related Zn finger transcription factor Osterix promotes proliferation of mesenchymal cells. Gene, 2006. 366(1): p. 145-151.
[183] Selvamurugan, N., et al., Parathyroid hormone regulates the rat collagenase-3 promoter in osteoblastic cells through the cooperative interaction of the activator protein-1 site and the runt domain binding sequence. Journal of Biological Chemistry, 1998. 273(17): p. 10647-10657.
[184] McCarthy, T.L., et al., Runt domain factor (Runx)-dependent effects on CCAAT/ enhancer-binding protein delta expression and activity in osteoblasts. J Biol Chem, 2000. 275(28): p. 21746-53.
[185] Gutierrez, S., et al., CCAAT/enhancer-binding proteins (C/EBP) beta and delta activate osteocalcin gene transcription and synergize with Runx2 at the C/EBP element to regulate bone-specific expression. Journal of Biological Chemistry, 2002. 277(2): p. 1316-1323.
[186] Xiao, G.Z., et al., Cooperative interactions between activating transcription factor 4 and Runx2/Cbfa1 stimulate osteoblast-specific osteocalcin gene expression. Journal of Biological Chemistry, 2005. 280(35): p. 30689-30696.
[187] Yang, X.G., et al., ATF4 is a substrate of RSK2 and an essential regulator of osteoblast biology: Implication for Coffin-Lowry syndrome. Cell, 2004. 117(3): p. 387-398.
[188] Otto, F., et al., Cbfa1, a candidate gene for cleidocranial dysplasia syndrome, is essential for osteoblast differentiation and bone development. Cell, 1997. 89(5): p. 765-771.
[189] Komori, T., Regulation of osteoblast differentiation by transcription factors. Journal of Cellular Biochemistry, 2006. 99(5): p. 1233-1239.
[190] Puchacz, E., et al., Chromosomal localization of the human osteocalcin gene. Endocrinology, 1989. 124(5): p. 2648-50.
[191] Cancela, L., et al., Molecular-structure, chromosome assignment, and promoter organization of the human matrix gla protein gene. Journal of Biological Chemistry, 1990. 265(25): p. 15040-15048.
[192] Lee, N.K., et al., Endocrine regulation of energy metabolism by the skeleton. Cell, 2007. 130(3): p. 456-469.
[193] Nomura, S., et al., Developmental expression of 2ar (osteopontin) and SPARC (osteonectin) RNA as revealed by in situ hybridization. J Cell Biol, 1988. 106(2): p. 441-50.
[194] Johansson C., The Structural Organization of Type IV Collagen. The journal of Biological Chemistry, 1992. 267(34): p. 24533-24537.
[195] Ashizawa, N., et al., Osteopontin is produced by rat cardiac fibroblasts and mediates A(II)-induced DNA synthesis and collagen gel contraction. Journal of Clinical Investigation, 1996. 98(10): p. 2218-2227.
[196] Murry, C.E., et al., MACROPHAGES EXPRESS OSTEOPONTIN DURING REPAIR OF MYOCARDIAL NECROSIS. American Journal of Pathology, 1994. 145(6): p. 1450-1462.
[197] Lkeda, T., et al., Osteopontin mRNA is expressed by smooth muscle-derived foam cells in human atherosclerotic lesions of the aorta. J Clin Invest 1993. 92(6): p. 2814-20.
[198] Uaesoontrachoon, K., et al., Osteopontin and skeletal muscle myoblasts: Association with muscle regeneration and regulation of myoblast function in vitro. International Journal of Biochemistry & Cell Biology, 2008. 40(10): p. 2303-2314.
[199] Merry, K., et al., Expression of osteopontin mRNA by osteoclasts and osteoblasts in modelling adult human bone. J Cell Sci, 1993. 104(Pt 4): p. 1013-20.
[200] Choi, S.T., et al., Osteopontin might be involved in bone remodelling rather than in inflammation in ankylosing spondylitis. Rheumatology, 2008. 47(12): p. 1775-1779.
[201] Reinholt, F.P., et al., Osteopontin - a possible anchor of osteoclasts to bone. Proceedings of the National Academy of Sciences of the United States of America, 1990. 87(12): p. 4473-4475.
[202] Solloway, M.J., et al., Molecular pathways in myocardial development: a stem cell perspective. Cardiovascular Research, 2003. 58(2): p. 264-77.
[203] Olson, E.N., et al., Molecular pathways controlling heart development. Science, 1996. 272(5262): p. 671-6.
[204] Arabadjiev, A., et al., We heart cultured hearts. A comparative review of methodologies for targeted differentiation and maintenance of cardiomyocytes derived from pluripotent and multipotent stem cells. BioDiscovery, 2014. 14(2)
[205] Smits, A.M., et al., Human cardiomyocyte progenitor cells differentiate into functional mature cardiomyocytes: an in vitro model for studying human cardiac physiology and pathophysiology. Nat Protoc., 2009. 4(2): p. 232-43.
[206] Fukuda. K., et al., Cardiomyocytes can be generated from marrow stromal cells in vitro. J. Clin. Invest., 1999. 103: p. 697-705.
[207] Shi S.T., et al., Differentiation of Bone Marrow Mesenchymal Stem Cells to Cardiomyocyte-Like Cells Is Regulated by the Combined Low Dose Treatment of Transforming Growth Factor-β1 and 5-Azacytidine. Stem Cells International, 2016. Article ID 3816256.
[208] Joupari M.D., et al., Effects of 5-Azacytidine on Differentiation of Ovine Mesenchymal Stem Cells. Int J Stem Cell Res Transplant., 2015. 03(2): p. 96-100.
[209] Martin-Rendon, E., et al., 5-Azacytidine-treated human mesenchymal stem/progenitor cells derived from umbilical cord, cord blood and bone marrow do not generate cardiomyocytes in vitro at high frequencies. Vox Sanguinis, 2008. 95(2): p. 137-148.
[210] Wang, H.Y., et al., Isolation and Characterization of Porcine Amniotic Fluid-Derived Multipotent Stem Cells. PLoS ONE, 2011. 6(5): e19964.
[211] Planat-Bénard, V., et al., Spontaneous Cardiomyocyte Differentiation From Adipose Tissue Stroma Cells. Circ Res., 2004. 94(2): p. 223-229.
[212] Okamoto, K., et al., ‘Working′ cardiomyocytes exhibiting plateau action potentials fromhumanplacenta-derived extraembryonic mesodermal cells. Experimental Cell Research, 2007. 313(12): p. 2550-2562.
[213] Xu, W.R., et al., Mesenchymal Stem Cells from Adult Human Bone Marrow Differentiate into a Cardiomyocyte Phenotype In Vitro. Experimental Biology and Medicine, 2004. 229(7): p. 623-31.
[214] Dimmeler, S., et al., Transdifferentiation of Blood-Derived Human Adult Endothelial Progenitor Cells Into Functionally Active Cardiomyocytes. Circulation, 2003. 107: p. 1024-1032.
[215] Pittenger, M.F., et al., Human Mesenchymal Stem Cells Differentiate to a Cardiomyocyte Phenotype in the Adult Murine Heart. Circulation, 2002.105: p. 93-98.
[216] Jacot, J.G., et al., Amniotic fluid-derived stem cells demonstrate limited cardiac differentiation following small molecule-based modulation of Wnt signaling pathway. Biomed. Mater., 2015. 10: 034103.
[217] Ravens, U., et al., 5-Azacytidine induces changes in electrophysiological properties of human mesenchymal stem cells. Cell Research, 2006. 16: p. 949-960.
[218] Kuo, S.M., et al., Hydrolyzed 5-Azacytidine Enhances Differentiation of Rat Mesenchymal Stem Cells into Cardiomyocytes. J. Med. Biol. Eng., 2015. 35: p. 473–481.
[219] Miaoli, M., et al., Amniotic fluid stem cells morph into a cardiovascular lineage: analysis of a chemically induced cardiac and vascular commitment. Drug Design, Development and Therapy, 2013. 7: p. 1063–1073.
[220] Mohanty, S., et al., TGFβ1 contributes to cardiomyogenic-like differentiation of human bone marrow mesenchymal stem cells. International Journal of Cardiology, 2013. 163(1): p. 93–99.
[221] Nikaido, T., et al., Human Amniotic Mesenchymal Cells Have Some Characteristics of Cardiomyocytes. Transplantation, 2005. 79: p. 528–535.
[222] Kim, B.S., et al., In vitro cardiomyogenic differentiation of adipose-derived stromal cells using transforming growth factor-b1. Cell Biochem Funct, 2009. 27: p. 148–154.
[223] Antonitsis, P., et al., In vitro cardiomyogenic differentiation of adult human bone marrow mesenchymal stem cells. The role of 5-azacytidine. Interactive CardioVascular and Thoracic Surgery, 2007. 6: p. 593–597.
[224] Guan, X., et al., In Vitro Cardiomyogenic Potential of Human Amniotic Fluid Stem Cells. J Tissue Eng Regen Med., 2011. 5(3): p. 220–228.
[225] Goumans, M.J., et al., TGF-beta1 induces efficient differentiation of human cardiomyocyte progenitor cells into functional cardiomyocytes in vitro. Stem Cell Res., 2007. 1: p. 138-49.
[226] Matsuura, K., et al., Adult cardiac Sca-1-positive cells differentiate into beating cardiomyocytes. J Biol Chem., 2004. 279(11): p. 384-91.
[227] Wang, H.S., et al., Mesenchymal stem cells in the Wharton′s jelly of the human umbilical cord. Stem Cells, 2004. 22: p. 1330-7.
[228] Wu, K.H., et al., Cardiac potential of stem cells from whole human umbilical cord tissue. J Cell Biochem., 2009. 107: p. 926-32.
[229] Jacot, J. G., et al., Amniotic fluid-derived stem cells demonstrated cardiogenic potential in indirect co-culture with human cardiac cells, Ann Biomed Eng., 2014. 42(12): p. 2490–2500.
[230] Yeh, Y. C., et al., Cellular Cardiomyoplasty with Human Amniotic Fluid Stem Cells: In Vitro and In Vivo Studies. Tissue Engineering: Part A, 2010. 16.
[231] Bollini, S., et al., In Vitro and In Vivo Cardiomyogenic Differentiation of Amniotic Fluid Stem Cells. Stem Cell Rev and Rep, 2011. 7: p. 364–380.
[232] Chua, K. H., et al., 5-Azacytidine Is Insufficient For Cardiogenesis In Human Adipose-Derived Stem Cells. Journal of Negative Results in BioMedicine, 2012. 11(3).
[233] Dou, Z. Y., et al., Effect of 5-azacytidine induction duration on differentiation of human first-trimester fetal mesenchymal stem cells towards cardiomyocyte-like cells. Interactive CardioVascular and Thoracic Surgery, 2009. 9: p. 943–946.
[234] Casteilla L., et al., Spontaneous Cardiomyocyte Differentiation From Adipose Tissue Stroma Cells. Circ Res., 2004. 94: p. 223-229.
[235] Chen, C.F., et al., Real-time quantification of microRNAs by stem-loop RT-PCR. Nucleic Acids Research, 2005. 33(20).
[236] Niesters, H.G.M., Quantitation of viral load using real-time amplification techniques. Methods, 2001. 25(4): p. 419-429.
[237] Livak, K.J. and T.D. Schmittgen, Analysis of relative gene expression data using real-time quantitative PCR and the 2(T) (-Delta Delta C) method. Methods, 2001. 25(4): p. 402-408.
[238] Schmittgen, T.D. and K.J. Livak, Analyzing real-time PCR data by the comparative C-T method. Nature Protocols, 2008. 3(6): p. 1101-1108.)
指導教授 樋口亞绀(Akon Higuchi) 審核日期 2016-7-18
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