參考文獻 |
1. Sada, A. and T. Tumbar, New Insights into Mechanisms of Stem Cell Daughter Fate Determination in Regenerative Tissues, in Intenational Review of Cell and Molecular Biology, Vol 300, K.W. Jeon, Editor. 2013. p. 1-50.
2. Zakrzewski, Stem cells: past, present, and future. Stem Cell Research & Therapy, 2019. 10.
3. Singh, V.K., Describing the Stem Cell Potency: The Various Methods of Functional Assessment and In silico Diagnostics. Frontiers in Cell and Developmental Biology, 2016. 4.
4. Erceg Ivkošić, I., Unlocking the Potential of Mesenchymal Stem Cells in Gynecology: Where Are We Now? Journal of personalized medicine, 2023. 13(8): p. 1253.
5. Boroviak, T. and J. Nichols, The birth of embryonic pluripotency. Philosophical Transactions of the Royal Society B: Biological Sciences, 2014. 369(1657): p. 20130541.
6. Chang, E.A., Human Induced Pluripotent Stem Cells : Clinical Significance and Applications in Neurologic Diseases. Journal of Korean Neurosurgical Society, 2019. 62(5): p. 493-501.
7. Teo, A.K.K. and L. Vallier, Emerging use of stem cells in regenerative medicine. Biochemical Journal, 2010. 428: p. 11-23.
8. Eguizabal, C., Two decades of embryonic stem cells: a historical overview. Human Reproduction Open, 2019. 2019(1).
9. Niu, X., Is day 7 culture necessary for in vitro fertilization of cryopreserved/warmed human oocytes? Reproductive Biology and Endocrinology, 2020. 18: p. 1-4.
10. Alteri, A., Busting the myth of extended blastocyst culture until Day 7: Protocol for systematic review and meta-analysis. Medicine, 2020. 99(5): p. e18909.
11. Higuchi, A., Biomaterials for the feeder-free culture of human embryonic stem cells and induced pluripotent stem cells. Chemical reviews, 2011. 111(5): p. 3021-3035.
12. Sills, E.S., Identification and isolation of embryonic stem cells in reproductive endocrinology: theoretical protocols for conservation of human embryos derived from in vitro fertilization. Theoretical Biology and Medical Modelling, 2005. 2.
13. Heins, N., Derivation, characterization, and differentiation of human embryonic stem cells. Stem cells, 2004. 22(3): p. 367-376.
14. Lo, B. and L. Parham, Ethical issues in stem cell research. Endocrine reviews, 2009. 30(3): p. 204-213.
15. Hyun, I., The bioethics of stem cell research and therapy. The Journal of clinical investigation, 2010. 120(1): p. 71-75.
16. Landry, D.W. and H.A. Zucker, Embryonic death and the creation of human embryonic stem cells. The Journal of Clinical Investigation, 2004. 114(9): p. 1184-1186.
17. 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.
18. Takahashi, K., Induction of pluripotent stem cells from adult human fibroblasts by defined factors. cell, 2007. 131(5): p. 861-872.
19. Okita, K. and S. Yamanaka, Induced pluripotent stem cells: opportunities and challenges. Philosophical Transactions of the Royal Society B: Biological Sciences, 2011. 366(1575): p. 2198-2207.
20. Ye, L., C. Swingen, and J. Zhang, Induced pluripotent stem cells and their potential for basic and clinical sciences. Current cardiology reviews, 2013. 9(1): p. 63-72.
21. Bai, Q., Embryonic stem cells or induced pluripotent stem cells? A DNA integrity perspective. Current Gene Therapy, 2013. 13(2): p. 93-98.
22. Yamanaka, S., Induced pluripotent stem cells: past, present, and future. Cell stem cell, 2012. 10(6): p. 678-684.
23. Nath, S.C., L. Menendez, and I.F. Ben-Nun, Overcoming the Variability of iPSCs in the Manufacturing of Cell-Based Therapies. International Journal of Molecular Sciences, 2023. 24(23).
24. Taylor, C.J., E.M. Bolton, and J.A. Bradley, Immunological considerations for embryonic and induced pluripotent stem cell banking. Philosophical Transactions of the Royal Society B: Biological Sciences, 2011. 366(1575): p. 2312-2322.
25. Moradi, S., Research and therapy with induced pluripotent stem cells (iPSCs): social, legal, and ethical considerations. Stem cell research & therapy, 2019. 10: p. 1-13.
26. Tian, Z., Rationale and methodology of reprogramming for generation of induced pluripotent stem cells and induced neural progenitor cells. International journal of molecular sciences, 2016. 17(4): p. 594.
27. Mitsui, K., Viral vector-based innovative approaches to directly abolishing tumorigenic pluripotent stem cells for safer regenerative medicine. Molecular Therapy Methods & Clinical Development, 2017. 5: p. 51-58.
28. Mamaeva, A., Quality Control of Human Pluripotent Stem Cell Colonies by Computational Image Analysis Using Convolutional Neural Networks. International Journal of Molecular Sciences, 2023. 24(1).
29. Ren, Y., Feeder cells treated with ethanol can be used to maintain self‐renewal and pluripotency of human pluripotent stem cells. FEBS Open bio, 2023. 13(2): p. 279-292.
30. Higuchi, A., Design of polymeric materials for culturing human pluripotent stem cells: Progress toward feeder-free and xeno-free culturing. Progress in Polymer Science, 2014. 39(7): p. 1348-1374.
31. Allison, T.F., Assessment of established techniques to determine developmental and malignant potential of human pluripotent stem cells. Nature Communications, 2018. 9.
32. Krasnova, O.A., Prognostic Analysis of Human Pluripotent Stem Cells Based on Their Morphological Portrait and Expression of Pluripotent Markers. International Journal of Molecular Sciences, 2022. 23(21).
33. Maruotti, J., A simple and scalable process for the differentiation of retinal pigment epithelium from human pluripotent stem cells. Stem cells translational medicine, 2013. 2(5): p. 341-354.
34. Guillevic, O., A novel molecular and functional stemness signature assessing human cord blood-derived endothelial progenitor cell immaturity. PLoS One, 2016. 11(4): p. e0152993.
35. Hoang, D.M., Stem cell-based therapy for human diseases. Signal transduction and targeted therapy, 2022. 7(1): p. 1-41.
36. De Masi, C., Application of CRISPR/Cas9 to human-induced pluripotent stem cells: from gene editing to drug discovery. Human genomics, 2020. 14: p. 1-12.
37. Chun, Y.S., K. Byun, and B. Lee, Induced pluripotent stem cells and personalized medicine: current progress and future perspectives. Anatomy & cell biology, 2011. 44(4): p. 245.
38. Lee, H. and M.-Y. Son, Current challenges associated with the use of human induced pluripotent stem cell-derived organoids in regenerative medicine. International Journal of Stem Cells, 2021. 14(1): p. 9.
39. De Coppi, P., Isolation of amniotic stem cell lines with potential for therapy. Nature Biotechnology, 2007. 25(1): p. 100-106.
40. Rennie, K., Applications of Amniotic Membrane and Fluid in Stem Cell Biology and Regenerative Medicine. Stem Cells International, 2012. 2012.
41. Antonucci, I., Amniotic Fluid Stem Cells: A Novel Source for Modeling of Human Genetic Diseases. International Journal of Molecular Sciences, 2016. 17(4).
42. Walther, G., J. Gekas, and O.F. Bertrand, Amniotic Stem Cells for Cellular Cardiomyoplasty: Promises and Premises. Catheterization and Cardiovascular Interventions, 2009. 73(7): p. 917-924.
43. Alonaizan, R. and C. Carr, Cardiac regeneration following myocardial infarction: the need for regeneration and a review of cardiac stromal cell populations used for transplantation. Biochemical Society Transactions, 2022. 50(1): p. 269-281.
44. Khan, M.A., Global epidemiology of ischemic heart disease: results from the global burden of disease study. Cureus, 2020. 12(7).
45. Li, N., New insights into the role of exosomes in the heart after myocardial infarction. Journal of cardiovascular translational research, 2019. 12: p. 18-27.
46. Li, Z., Targeted anti–IL-1β platelet microparticles for cardiac detoxing and repair. Science advances, 2020. 6(6): p. eaay0589.
47. Huang, J., Human pluripotent stem cell-derived cardiac cells: application in disease modeling, cell therapy, and drug discovery. Frontiers in Cell and Developmental Biology, 2021. 9: p. 655161.
48. Yu, D., X. Wang, and L. Ye, Cardiac tissue engineering for the treatment of myocardial infarction. Journal of Cardiovascular Development and Disease, 2021. 8(11): p. 153.
49. Mummery, C.L., Differentiation of human embryonic stem cells and induced pluripotent stem cells to cardiomyocytes: a methods overview. Circulation research, 2012. 111(3): p. 344-358.
50. Balafkan, N., A method for differentiating human induced pluripotent stem cells toward functional cardiomyocytes in 96-well microplates. Scientific Reports, 2020. 10(1).
51. D′Amour, K.A., Efficient differentiation of human embryonic stem cells to definitive endoderm. Nature biotechnology, 2005. 23(12): p. 1534-1541.
52. Kehat, I., Human embryonic stem cells can differentiate into myocytes with structural and functional properties of cardiomyocytes. The Journal of clinical investigation, 2001. 108(3): p. 407-414.
53. Mummery, C., Differentiation of human embryonic stem cells to cardiomyocytes: role of coculture with visceral endoderm-like cells. Circulation, 2003. 107(21): p. 2733-2740.
54. Passier, R., Increased cardiomyocyte differentiation from human embryonic stem cells in serum‐free cultures. Stem Cells, 2005. 23(6): p. 772-780.
55. Kishimoto, K., Directed differentiation of human pluripotent stem cells into diverse organ-specific mesenchyme of the digestive and respiratory systems. Nature protocols, 2022. 17(11): p. 2699-2719.
56. Bhattacharya, S., High efficiency differentiation of human pluripotent stem cells to cardiomyocytes and characterization by flow cytometry. JoVE (Journal of Visualized Experiments), 2014(91): p. e52010.
57. Shao, Y., J.M. Sang, and J.P. Fu, On human pluripotent stem cell control: The rise of 3D bioengineering and mechanobiology. Biomaterials, 2015. 52: p. 26-43.
58. Xu, Y.B., Biomaterials for stem cell engineering and biomanufacturing. Bioactive Materials, 2019. 4: p. 366-379.
59. Park, Y.-G., Effects of feeder cell types on culture of mouse embryonic stem cell in vitro. Development & reproduction, 2015. 19(3): p. 119.
60. Kropp, C., D. Massai, and R. Zweigerdt, Progress and challenges in large-scale expansion of human pluripotent stem cells. Process Biochemistry, 2017. 59: p. 244-254.
61. Meng, G., A novel method for generating xeno-free human feeder cells for human embryonic stem cell culture. Stem cells and development, 2008. 17(3): p. 413-422.
62. Zou, Q., Development of a Xeno-Free Feeder-Layer System from Human Umbilical Cord Mesenchymal Stem Cells for Prolonged Expansion of Human Induced Pluripotent Stem Cells in Culture. Plos One, 2016. 11(2).
63. Higuchi, A., Long-term xeno-free culture of human pluripotent stem cells on hydrogels with optimal elasticity. Scientific Reports, 2015. 5.
64. Kawase, E. and N. Nakatsuji, Development of substrates for the culture of human pluripotent stem cells. Biomaterials Science, 2023. 11(9): p. 2974-2987.
65. Higuchi, A., Biomimetic cell culture proteins as extracellular matrices for stem cell differentiation. Chemical reviews, 2012. 112(8): p. 4507-4540.
66. Aisenbrey, E.A. and W.L. Murphy, Synthetic alternatives to Matrigel. Nature Reviews Materials, 2020. 5(7): p. 539-551.
67. Vukicevic, S., Identification of multiple active growth factors in basement membrane Matrigel suggests caution in interpretation of cellular activity related to extracellular matrix components. Experimental cell research, 1992. 202(1): p. 1-8.
68. Salo, T., A novel human leiomyoma tissue derived matrix for cell culture studies. BMC cancer, 2015. 15: p. 1-16.
69. Ahmadian Baghbaderani, B., A newly defined and xeno-free culture medium supports every-other-day medium replacement in the generation and long-term cultivation of human pluripotent stem cells. PLoS One, 2016. 11(9): p. e0161229.
70. Bandzerewicz, A. and A. Gadomska-Gajadhur, Into the tissues: Extracellular matrix and its artificial substitutes: Cell signalling mechanisms. Cells, 2022. 11(5): p. 914.
71. Karamanos, N.K., A guide to the composition and functions of the extracellular matrix. The FEBS journal, 2021. 288(24): p. 6850-6912.
72. Yue, B., Biology of the extracellular matrix: an overview. Journal of glaucoma, 2014. 23: p. S20-S23.
73. Chen, G., Chemically defined conditions for human iPSC derivation and culture. Nature methods, 2011. 8(5): p. 424-429.
74. Rodin, S., Long-term self-renewal of human pluripotent stem cells on human recombinant laminin-511. Nature biotechnology, 2010. 28(6): p. 611.
75. Dzobo, K. and C. Dandara, The extracellular matrix: its composition, function, remodeling, and role in tumorigenesis. Biomimetics, 2023. 8(2): p. 146.
76. Kular, J.K., S. Basu, and R.I. Sharma, The extracellular matrix: Structure, composition, age-related differences, tools for analysis and applications for tissue engineering. Journal of tissue engineering, 2014. 5: p. 2041731414557112.
77. Krebsbach, P.H. and L.G. Villa-Diaz, The Role of Integrin α6 (CD49f) in Stem Cells: More than a Conserved Biomarker. Stem Cells and Development, 2017. 26(15): p. 1090-1099.
78. Pang, X.C., Targeting integrin pathways: mechanisms and advances in therapy. Signal Transduction and Targeted Therapy, 2023. 8(1).
79. Xu, C.H., Feeder-free growth of undifferentiated human embryonic stem cells. Nature Biotechnology, 2001. 19(10): p. 971-974.
80. Mezu-Ndubuisi, O.J. and A. Maheshwari, The role of integrins in inflammation and angiogenesis. Pediatric Research, 2021. 89(7): p. 1619-1626.
81. Goswami, S., Importance of integrin receptors in the field of pharmaceutical & medical science. 2013.
82. Jia, J., Development of peptide-functionalized synthetic hydrogel microarrays for stem cell and tissue engineering applications. Acta Biomaterialia, 2016. 45: p. 110-120.
83. Chen, Y.M., Xeno-free culture of human pluripotent stem cells on oligopeptide-grafted hydrogels with various molecular designs. Scientific Reports, 2017. 7.
84. Hayashi, Y. and M.K. Furue, Biological effects of culture substrates on human pluripotent stem cells. Stem cells international, 2016. 2016.
85. Yang, K., J. Lee, and S.-W. Cho, Engineering biomaterials for feeder-free maintenance of human pluripotent stem cells. International journal of stem cells, 2012. 5(1): p. 1.
86. Sung, T.-C., Design of dual peptide-conjugated hydrogels for proliferation and differentiation of human pluripotent stem cells. Materials Today Bio, 2024. 25: p. 100969.
87. Hozumi, K. and M. Nomizu, Mixed peptide-conjugated chitosan matrices as multi-receptor targeted cell-adhesive scaffolds. International Journal of Molecular Sciences, 2018. 19(9): p. 2713.
88. Musilkova, J., Cell adhesion and growth enabled by biomimetic oligopeptide modification of a polydopamine-poly (ethylene oxide) protein repulsive surface. Journal of Materials Science: Materials in Medicine, 2015. 26: p. 1-13.
89. 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.
90. Melkoumian, Z., 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.
91. Cooke, M., Neural differentiation regulated by biomimetic surfaces presenting motifs of extracellular matrix proteins. Journal of Biomedical Materials Research Part A: An Official Journal of The Society for Biomaterials, The Japanese Society for Biomaterials, and The Australian Society for Biomaterials and the Korean Society for Biomaterials, 2010. 93(3): p. 824-832.
92. Nomizu, M., Cell binding sequences in mouse laminin α1 chain. Journal of Biological Chemistry, 1998. 273(49): p. 32491-32499.
93. Suzuki, S., 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.
94. Higuchi, A., Polymeric materials for ex vivo expansion of hematopoietic progenitor and stem cells. Journal of Macromolecular Science®, Part C: Polymer Reviews, 2009. 49(3): p. 181-200.
95. Oldberg, A., Identification of a bone sialoprotein receptor in osteosarcoma cells. Journal of Biological Chemistry, 1988. 263(36): p. 19433-19436.
96. Salasznyk, R.M., Adhesion to vitronectin and collagen I promotes osteogenic differentiation of human mesenchymal stem cells. BioMed Research International, 2004. 2004: p. 24-34.
97. Kondo, M., Lymphoid and myeloid lineage commitment in multipotent hematopoietic progenitors. Immunological reviews, 2010. 238(1): p. 37-46.
98. Wu, L. and Y.-J. Liu, Development of dendritic-cell lineages. Immunity, 2007. 26(6): p. 741-750.
99. Hassan, G. and M. Seno, Blood and cancer: cancer stem cells as origin of hematopoietic cells in solid tumor microenvironments. Cells, 2020. 9(5): p. 1293.
100. Davila, J.A.A. and A.H. De Los Rios, An overview of peripheral blood mononuclear cells as a model for immunological research of Toxoplasma gondii and other apicomplexan parasites. Frontiers in cellular and infection microbiology, 2019. 9.
101. Corkum, C.P., Immune cell subsets and their gene expression profiles from human PBMC isolated by Vacutainer Cell Preparation Tube (CPT™) and standard density gradient. BMC immunology, 2015. 16: p. 1-18.
102. Wood, K.J., F. Issa, and J. Hester, Understanding stem cell immunogenicity in therapeutic applications. Trends in immunology, 2016. 37(1): p. 5-16.
103. Ye, Q., Generation of universal and hypoimmunogenic human pluripotent stem cells. Cell proliferation, 2020. 53(12): p. e12946.
104. Wang, D., Targeted disruption of the β 2-microglobulin gene minimizes the immunogenicity of human embryonic stem cells. Stem cells translational medicine, 2015. 4(10): p. 1234-1245.
105. Deuse, T., Hypoimmunogenic derivatives of induced pluripotent stem cells evade immune rejection in fully immunocompetent allogeneic recipients. Nature biotechnology, 2019. 37(3): p. 252-258.
106. Jang, Y., Development of immunocompatible pluripotent stem cells via CRISPR-based human leukocyte antigen engineering. Experimental & Molecular Medicine, 2019. 51(1): p. 1-11.
107. Torikai, H., Toward eliminating HLA class I expression to generate universal cells from allogeneic donors. Blood, The Journal of the American Society of Hematology, 2013. 122(8): p. 1341-1349.
108. Zhao, H.-X., Enhanced immunological tolerance by HLA-G1 from neural progenitor cells (NPCs) derived from human embryonic stem cells (hESCs). Cellular Physiology and Biochemistry, 2017. 44(4): p. 1435-1444.
109. Rong, Z., An effective approach to prevent immune rejection of human ESC-derived allografts. Cell stem cell, 2014. 14(1): p. 121-130.
110. Sung, T.C., Design of dual peptide-conjugated hydrogels for proliferation and differentiation of human pluripotent stem cells. Materials Today Bio, 2024. 25.
111. Sung, T.C., Poly(vinyl alcohol-<i>co</i>-itaconic acid) hydrogels grafted with several designed peptides for human pluripotent stem cell culture and differentiation into cardiomyocytes. Journal of Materials Chemistry B, 2021. 9(37): p. 7662-7673.
112. Sung, T.C., Cell-binding peptides on the material surface guide stem cell fate of adhesion, proliferation and differentiation. Journal of Materials Chemistry B, 2023. 11(7): p. 1389-1415. |