[1] Jedrzejczak-Silicka, M. History of cell culture. in New Insights into Cell Culture Technology (ed Gowder, S. J. T.) (IntechOpen, London. 2017). doi: 10.5772/66905
[2] Elsdale, T. & Bard, J. Collagen substrata for studies on cell behavior. Journal of Cell Biology 54, 626-637 (1972). doi: 10.1083/jcb.54.3.626
[3] Thiele, J. et al. 25th anniversary article: designer hydrogels for cell cultures: a materials selection guide. Advanced Materials 26, 125-147 (2014). doi: 10.1002/adma.201302958
[4] Reed, J. et al. In situ mechanical interferometry of matrigel films. Langmuir 25, 36-39 (2009). doi: 10.1021/la8033098
[5] Orkin, R. W. et al. A murine tumor producing a matrix of basement membrane. The Journal of Experimental Medicine 145, 204-220 (1977). doi: 10.1084/jem.145.1.204
[6] Benton, G. et al. Matrigel: from discovery and ECM mimicry to assays and models for cancer research. Advanced Drug Delivery Reviews 79-80, 3-18 (2014). doi: 10.1016/j.addr.2014.06.005
[7] Kwee, B. J. & Mooney, D. J. Biomaterials for skeletal muscle tissue engineering. Current Opinion in Biotechnology 47, 16-22 (2017). doi: 10.1016/j.copbio.2017.05.003
[8] Moroni, L. et al. Biofabrication: a guide to technology and terminology. Trends in Biotechnology 36, 384-402 (2018). doi: 10.1016/j.tibtech.2017.10.015
[9] Loebel, C. & Burdick, J. A. Engineering stem and stromal cell therapies for musculoskeletal tissue repair. Cell Stem Cell 22, 325-339 (2018). doi: 10.1016/j.stem.2018.01.014
[10] Moroni, L. et al. Biofabrication strategies for 3D in vitro models and regenerative medicine. Nature Reviews Materials 3, 21-37 (2018). doi: 10.1038/s41578-018-0006-y
[11] Juliano, R. L. & Haskill, S. Signal transduction from the extracellular matrix. The Journal of Cell Biology 120, 577-585 (1993). doi: 10.1083/jcb.120.3.577
[12] Pelham, R. J. Jr. & Wang, Y. L. Cell locomotion and focal adhesions are regulated by substrate flexibility. Proceedings of the National Academy of Sciences of the United States of America 94, 13661-13665 (1997). doi: 10.1073/pnas.94.25.13661
[13] Chaudhuri, O. et al. Effects of extracellular matrix viscoelasticity on cellular behaviour. Nature 584, 535-546 (2020). doi: 10.1038/s41586-020-2612-2
[14] Hadden, W. J. et al. Stem cell migration and mechanotransduction on linear stiffness gradient hydrogels. Proceedings of the National Academy of Sciences of the United States of America 114, 5647-5652 (2017). doi: 10.1073/pnas.1618239114
[15] Cosgrove, B. D. et al. N-cadherin adhesive interactions modulate matrix mechanosensing and fate commitment of mesenchymal stem cells. Nature Materials 15, 1297-1306 (2016). doi: 10.1038/nmat4725
[16] Darnell, M. et al. Material microenvironmental properties couple to induce distinct transcriptional programs in mammalian stem cells. Proceedings of the National Academy of Sciences of the United States of America 115, E8368-E8377 (2018). doi: 10.1073/pnas.1802568115
[17] Engler, A. J. et al. Matrix elasticity directs stem cell lineage specification. Cell 126, 677-689 (2006). doi: 10.1016/j.cell.2006.06.044
[18] Soofi, S. S. et al. The elastic modulus of MatrigelTM as determined by atomic force microscopy. Journal of Structural Biology 167, 216-219 (2009). doi: 10.1016/j.jsb.2009.05.005
[19] Lou, J. Z. et al. Stress relaxing hyaluronic acid-collagen hydrogels promote cell spreading, fiber remodeling, and focal adhesion formation in 3D cell culture. Biomaterials 154, 213-222 (2018). doi: 10.1016/j.biomaterials.2017.11.004
[20] Chaudhuri, O. et al. Hydrogels with tunable stress relaxation regulate stem cell fate and activity. Nature Materials 15, 326-334 (2016). doi: 10.1038/nmat4489
[21] Garreta, E. et al. Fine tuning the extracellular environment accelerates the derivation of kidney organoids from human pluripotent stem cells. Nature Materials 18, 397-405 (2019). doi: 10.1038/s41563-019-0287-6
[22] Li, Y. W. et al. Volumetric compression induces intracellular crowding to control intestinal organoid growth via wnt/β-catenin signaling. Cell Stem Cell 28, 63-78.e7 (2021). doi: 10.1016/j.stem.2020.09.012
[23] Lawlor, K. T. et al. Cellular extrusion bioprinting improves kidney organoid reproducibility and conformation. Nature Materials, 20, 260-271 (2021). doi: 10.1038/s41563-020-00853-9
[24] Chiou, G. et al. Scaffold architecture and matrix strain modulate mesenchymal cell and microvascular growth and development in a time dependent manner. Cellular and Molecular Bioengineering 13, 507-526 (2020). doi: 10.1007/s12195-020-00648-7
[25] Mondal, G., Barui, S. & Chaudhuri, A. The relationship between the cyclic-RGDfK ligand and αvβ3 integrin receptor. Biomaterials 34, 6249-6260 (2013). doi: 10.1016/j.biomaterials.2013.04.065
[26] Lambshead, J. W. et al. Long-term maintenance of human pluripotent stem cells on cRGDfK-presenting synthetic surfaces. Scientific Reports 8, 701 (2018). doi: 10.1038/s41598-018-19209-0
[27] Farach-Carson, M. C. & Carson, D. D. Perlecan—a multifunctional extracellular proteoglycan scaffold. Glycobiology 17, 897-905 (2007). doi: 10.1093/glycob/cwm043
[28] Furue, M. K. et al. Heparin promotes the growth of human embryonic stem cells in a defined serum-free medium. Proceedings of the National Academy of Sciences of the United States of America 105, 13409-13414 (2008). doi: 10.1073/pnas.0806136105
[29] Klim, J. R. et al. A defined glycosaminoglycan-binding substratum for human pluripotent stem cells. Nature Methods 7, 989-994 (2010). doi: 10.1038/nmeth.1532
[30] Musah, S. et al. Glycosaminoglycan-binding hydrogels enable mechanical control of human pluripotent stem cell self-renewal. ACS Nano 6, 10168-10177 (2012). doi: 10.1021/nn3039148
[31] Nowak, M. et al. Modular GAG-matrices to promote mammary epithelial morphogenesis in vitro. Biomaterials 112, 20-30 (2017). doi: 10.1016/j.biomaterials.2016.10.007
[32] Juliar, B. A. et al. Cell-mediated matrix stiffening accompanies capillary morphogenesis in ultra-soft amorphous hydrogels. Biomaterials 230, 119634 (2020). doi: 10.1016/j.biomaterials.2019.119634
[33] Khademhosseini, A. & Langer, R. A decade of progress in tissue engineering. Nature Protocols 11, 1775-1781 (2016). doi: 10.1038/nprot.2016.123
[34] Berthiaume, F., Maguire, T. J. & Yarmush, M. L. Tissue engineering and regenerative medicine: history, progress, and challenges. Annual Review of Chemical and Biomolecular Engineering 2, 403-430 (2011). doi: 10.1146/annurev-chembioeng-061010-114257
[35] Yin, X. L. et al. Engineering stem cell organoids. Cell Stem Cell 18, 25-38 (2016). doi: 10.1016/j.stem.2015.12.005
[36] Lancaster, M. A. & Knoblich, J. A. Organogenesis in a dish: modeling development and disease using organoid technologies. Science 345, 1247125 (2014). doi: 10.1126/science.1247125
[37] Echave, M. C. et al. Gelatin as biomaterial for tissue engineering. Current Pharmaceutical Design 23, 3567-3584 (2017). doi: 10.2174/0929867324666170511123101
[38] Ma, S. H. et al. Gel microrods for 3D tissue printing. Advanced Biosystems 1, 1700075 (2017). doi: 10.1002/adbi.201700075
[39] Nichol, J. W. et al. Cell-laden microengineered gelatin methacrylate hydrogels. Biomaterials 31, 5536-5544 (2010). doi: 10.1016/j.biomaterials.2010.03.064
[40] Kolesky, D. B. et al. Three-dimensional bioprinting of thick vascularized tissues. Proceedings of the National Academy of Sciences of the United States of America 113, 3179-3184 (2016). doi: 10.1073/pnas.1521342113
[41] Oryan, A. et al. Chemical crosslinking of biopolymeric scaffolds: current knowledge and future directions of crosslinked engineered bone scaffolds. International Journal of Biological Macromolecules 107, 678-688 (2018). doi: 10.1016/j.ijbiomac.2017.08.184
[42] Gattazzo, F. et al. Gelatin-genipin-based biomaterials for skeletal muscle tissue engineering. Journal of Biomedical Materials Research Part B: Applied Biomaterials 106, 2763-2777 (2018). doi: 10.1002/jbm.b.34057
[43] Kitagawa, T. et al. Three-dimensional cell seeding and growth in radial-flow perfusion bioreactor for in vitro tissue reconstruction. Biotechnology and Bioengineering 93, 947-954 (2006). doi: 10.1002/bit.20797
[44] Olivares, A. L. & Lacroix, D. Simulation of cell seeding within a three-dimensional porous scaffold: a fluid-particle analysis. Tissue Engineering Part C: Methods 18, 624-631 (2012). doi: 10.1089/ten.tec.2011.0660
[45] Mao, D. et al. Bio-orthogonal click reaction-enabled highly specific in situ cellularization of tissue engineering scaffolds. Biomaterials 230, 119615 (2020). doi: 10.1016/j.biomaterials.2019.119615
[46] Bidarra, S. J., Barrias, C. C. & Granja, P. L. Injectable alginate hydrogels for cell delivery in tissue engineering. Acta Biomaterialia 10, 1646-1662 (2014). doi: 10.1016/j.actbio.2013.12.006
[47] Kuo, C. K. & Ma, P. X. Ionically crosslinked alginate hydrogels as scaffolds for tissue engineering: part 1. Structure, gelation rate and mechanical properties. Biomaterials 22, 511-521 (2001). doi: 10.1016/S0142-9612(00)00201-5
[48] Tibbits, S. 4D printing: multi‐material shape change. Architectural Design 84, 116-121 (2014). doi: 10.1002/ad.1710
[49] Galarraga, J. H. & Burdick, J. A. Moving hydrogels to the fourth dimension. Nature Materials 18, 914-915 (2019). doi: 10.1038/s41563-019-0458-5
[50] Burdick, J. A. & Murphy, W. L. Moving from static to dynamic complexity in hydrogel design. Nature Communications 3, 1269 (2012). doi: 10.1038/ncomms2271
[51] Khetan, S. et al. Degradation-mediated cellular traction directs stem cell fate in covalently crosslinked three-dimensional hydrogels. Nature Materials 12, 458-465 (2013). doi: 10.1038/nmat3586
[52] Caiazzo, M. et al. Defined three-dimensional microenvironments boost induction of pluripotency. Nature Materials 15, 344-352 (2016). doi: 10.1038/nmat4536
[53] Purcell, B. P. et al. Injectable and bioresponsive hydrogels for on-demand matrix metalloproteinase inhibition. Nature Materials 13, 653-661 (2014). doi: 10.1038/nmat3922
[54] Ye, H. Y. et al. Polyester elastomers for soft tissue engineering. Chemical Society Reviews 47, 4545-4580 (2018). doi: 10.1039/C8CS00161H
[55] Matsunaga, Y. T., Morimoto, Y. & Takeuchi, S. Molding cell beads for rapid construction of macroscopic 3D tissue architecture. Advanced Materials 23, H90-H94 (2011). doi: 10.1002/adma.201004375
[56] Morimoto, Y., Tanaka, R. & Takeuchi, S. Construction of 3D, layered skin, microsized tissues by using cell beads for cellular function analysis. Advanced Healthcare Materials 2, 261-265 (2013). doi: 10.1002/adhm.201200189
[57] Wen, Y. J. et al. Spinal cord injury repair by implantation of structured hyaluronic acid scaffold with PLGA microspheres in the rat. Cell and Tissue Research 364, 17-28 (2016). doi: 10.1007/s00441-015-2298-1
[58] Zhao, H. R. et al. Microfluidic synthesis of injectable angiogenic microgels. Cell Reports Physical Science 1, 100047 (2020). doi: 10.1016/j.xcrp.2020.100047
[59] Ma, S. H. Microfluidics tubing as a synthesizer for ordered microgel networks. Soft Matter 15, 3848-3853 (2019). doi: 10.1039/C9SM00626E
[60] Shang, L. R., Cheng, Y. & Zhao, Y. J. Emerging droplet microfluidics. Emerging droplet microfluidics. Chemical Reviews 117, 7964-8040 (2017). doi: 10.1021/acs.chemrev.6b00848
[61] Li, W. et al. Microfluidic fabrication of microparticles for biomedical applications. Chemical Society Reviews 47, 5646-5683 (2018). doi: 10.1039/C7CS00263G
[62] Ma, S. et al. Monodisperse collagen-gelatin beads as potential platforms for 3D cell culturing. Journal of Materials Chemistry. B 1, 5128-5136 (2013). doi: 10.1039/c3tb20851f
[63] Wang, W. et al. Hole-shell microparticles from controllably evolved double emulsions. Angewandte Chemie International Edition 52, 8084-8087 (2013). doi: 10.1002/anie.201301590
[64] Deng, N. N. et al. Wetting-induced formation of controllable monodisperse multiple emulsions in microfluidics. Lab on a Chip 13, 4047-4052 (2013). doi: 10.1039/C3LC50638J
[65] Ma, S. H. et al. Fabrication of microgel particles with complex shape via selective polymerization of aqueous two-phase systems. Small 8, 2356-2360 (2012). doi: 10.1002/smll.201102715
[66] Chu, L. Y. et al. Controllable monodisperse multiple emulsions. Angewandte Chemie International Edition 46, 8970-8974 (2007). doi: 10.1002/anie.200701358
[67] Ma, S. H., Huck, W. T. S. & Balabani, S. Deformation of double emulsions under conditions of flow cytometry hydrodynamic focusing. Lab on a Chip 15, 4291-4301 (2015). doi: 10.1039/C5LC00693G
[68] Ma, S. H. et al. On the flow topology inside droplets moving in rectangular microchannels. Lab on a Chip 14, 3611-3620 (2014). doi: 10.1039/C4LC00671B
[69] Ma, S. H. et al. The microenvironment of double emulsions in rectangular microchannels. Lab on a Chip 15, 2327-2334 (2015). doi: 10.1039/C5LC00346F
[70] Sorushanova, A. et al. The collagen suprafamily: from biosynthesis to advanced biomaterial development. Advanced Materials 31, 1801651 (2019). doi: 10.1002/adma.201801651
[71] Singh, P., Carraher, C. & Schwarzbauer, J. E. Assembly of fibronectin extracellular matrix. Annual Review of Cell and Developmental Biology 26, 397-419 (2010). doi: 10.1146/annurev-cellbio-100109-104020
[72] Bonnans, C., Chou, J. & Werb, Z. Remodelling the extracellular matrix in development and disease. Nature Reviews Molecular Cell Biology 15, 786-801 (2014). doi: 10.1038/nrm3904
[73] Onoe, H. et al. Metre-long cell-laden microfibres exhibit tissue morphologies and functions. Nature Materials 12, 584-590 (2013). doi: 10.1038/nmat3606
[74] Kang, E. et al. Digitally tunable physicochemical coding of material composition and topography in continuous microfibres. Nature Materials 10, 877-883 (2011). doi: 10.1038/nmat3108
[75] Xin, G. Q. et al. Microfluidics-enabled orientation and microstructure control of macroscopic graphene fibres. Nature Nanotechnology 14, 168-175 (2019). doi: 10.1038/s41565-018-0330-9
[76] Cao, Y. X. et al. Cascade pumping overcomes hydraulic resistance and moderates shear conditions for slow gelatin fiber shaping in narrow tubes. iScience 23, 101228 (2020). doi: 10.1016/j.isci.2020.101228
[77] An, D. et al. Designing a retrievable and scalable cell encapsulation device for potential treatment of type 1 diabetes. Proceedings of the National Academy of Sciences of the United States of America 115, E263-E272 (2018). doi: 10.1073/pnas.1708806115
[78] Jun, Y. et al. Microfluidics-generated pancreatic islet microfibers for enhanced immunoprotection. Biomaterials 34, 8122-8130 (2013). doi: 10.1016/j.biomaterials.2013.07.079
[79] Bhattacharjee, T. et al. Writing in the granular gel medium. Science Advances 1, e1500655 (2015). doi: 10.1126/sciadv.1500655
[80] Lee, A. et al. 3D bioprinting of collagen to rebuild components of the human heart. Science 365, 482-487 (2019). doi: 10.1126/science.aav9051
[81] Kunze, A. et al. Micropatterning neural cell cultures in 3D with a multi-layered scaffold. Biomaterials 32, 2088-2098 (2011). doi: 10.1016/j.biomaterials.2010.11.047
[82] Muerza-Cascante, M. L. et al. Melt electrospinning and its technologization in tissue engineering. Tissue Engineering Part B: Reviews 21, 187-202 (2015). doi: 10.1089/ten.teb.2014.0347
[83] Hong, J. et al. Cell-electrospinning and its application for tissue engineering. International Journal of Molecular Sciences 20, 6208 (2019). doi: 10.3390/ijms20246208
[84] Murphy, S. V. & Atala, A. 3D bioprinting of tissues and organs. Nature Biotechnology 32, 773-785 (2014). doi: 10.1038/nbt.2958
[85] Murphy, S. V., De Coppi, P. & Atala, A. Opportunities and challenges of translational 3D bioprinting. Nature Biomedical Engineering 4, 370-380 (2020). doi: 10.1038/s41551-019-0471-7
[86] Kang, H. W. et al. A 3D bioprinting system to produce human-scale tissue constructs with structural integrity. Nature Biotechnology 34, 312-319 (2016). doi: 10.1038/nbt.3413
[87] Ma, X. Y. et al. Deterministically patterned biomimetic human iPSC-derived hepatic model via rapid 3D bioprinting. Proceedings of the National Academy of Sciences of the United States of America 113, 2206-2211 (2016). doi: 10.1073/pnas.1524510113
[88] Grigoryan, B. et al. Multivascular networks and functional intravascular topologies within biocompatible hydrogels. Science 364, 458-464 (2019). doi: 10.1126/science.aav9750
[89] Bajaj, P. et al. 3D biofabrication strategies for tissue engineering and regenerative medicine. Annual Review of Biomedical Engineering 16, 247-276 (2014). doi: 10.1146/annurev-bioeng-071813-105155
[90] Soman, P. et al. Digital microfabrication of user-defined 3D microstructures in cell-laden hydrogels. Biotechnology and Bioengineering 110, 3038-3047 (2013). doi: 10.1002/bit.24957
[91] Hribar, K. C. et al. Light-assisted direct-write of 3D functional biomaterials. Lab on a Chip 14, 268-275 (2014). doi: 10.1039/C3LC50634G
[92] Melchels, F. P., Feijen, J. & Grijpma, D. W. A review on stereolithography and its applications in biomedical engineering. Biomaterials 31, 6121-6130 (2010). doi: 10.1016/j.biomaterials.2010.04.050
[93] Bhattacharjee, N. et al. Desktop-stereolithography 3D-Printing of a poly(dimethylsiloxane)-based material with sylgard-184 properties. Advanced Materials 30, 1800001 (2018). doi: 10.1002/adma.201800001
[94] Bhatia, S. N. et al. Effect of cell-cell interactions in preservation of cellular phenotype: cocultivation of hepatocytes and nonparenchymal cells. The FASEB Journal 13, 1883-1900 (1999). doi: 10.1096/fasebj.13.14.1883
[95] Li, X. D. et al. Inkjet bioprinting of biomaterials. Chemical Reviews 120, 10793-10833 (2020). doi: 10.1021/acs.chemrev.0c00008