Advances in precise cellmanipulation

Yuan Ma, Zhenwei Liang, Yiqing Chen, Jiadao Wang

Droplet ›› 2025, Vol. 4 ›› Issue (1) : e149.

PDF
Droplet ›› 2025, Vol. 4 ›› Issue (1) : e149. DOI: 10.1002/dro2.149
REVIEW ARTICLE

Advances in precise cellmanipulation

Author information +
History +

Abstract

Research on cells and organ-like tissues is critical in the fields of molecular biology, genetic analysis, proteomics analysis, tissue engineering, and others. In recent years, advancements in precise cell manipulation technologies have made precise positioning and batch processing of cells feasible. Various methods are used for cell recognition, positioning, manipulation, and assembly, often introducing external fields such as electric, magnetic, acoustic, or optical fields into the liquid environment to interact with cells, applying forces to induce cell movement and rearrangement. Alternatively, three-dimensional (3D) bioprinting technology is employed for precise cell positioning and assembly. This review will comprehensively assess the status, principles, advantages, disadvantages, and prospects of these precise cell manipulation technologies, covering single-cell manipulation, multicellular assembly, and biological 3D printing techniques.

Cite this article

Download citation ▾
Yuan Ma, Zhenwei Liang, Yiqing Chen, Jiadao Wang. Advances in precise cellmanipulation. Droplet, 2025, 4(1): e149 https://doi.org/10.1002/dro2.149

References

[1]
Gao D, Jin F, Zhou M, Jiang Y. Recent advances in single cell manipulation and biochemical analysis on microfluidics. Analyst. 2019;144:766-781.
CrossRef Google scholar
[2]
Yun H, Kim K, Lee WG. Cell manipulation in microfluidics. Biofabrication. 2013;5:022001.
CrossRef Google scholar
[3]
Dippong M, Carl P, Lenz C, et al. Hapten-specific single-cell selection of hybridoma clones by fluorescence-activated cell sorting for the generation of monoclonal antibodies. Anal Chem. 2017;89:4007-4012.
CrossRef Google scholar
[4]
Guillotin B, Guillemot F. Cell patterning technologies for organotypic tissue fabrication. Trends Biotechnol. 2011;29:183-190.
CrossRef Google scholar
[5]
Besant JD, Mohamadi RM, Aldridge PM, et al. Velocity valleys enable efficient capture and spatial sorting of nanoparticle-bound cancer cells. Nanoscale. 2015;7:6278-6285.
CrossRef Google scholar
[6]
Ma Y, Chen K, Xia F, et al. Phage-based profiling of rare single cells using nanoparticle-directed capture. ACS Nano. 2021;15:19202-19210.
CrossRef Google scholar
[7]
Poudineh M, Aldridge PM, Ahmed S, et al. Tracking the dynamics of circulating tumour cell phenotypes using nanoparticle-mediated magnetic ranking. Nat Nanotechnol. 2017;12:274-281.
CrossRef Google scholar
[8]
Ren T, Maitusong M, Zhou X, et al. Programing cell assembly via ink-free, label-free Magneto-Archimedes based strategy. ACS Nano. 2023;17:12072-12086.
CrossRef Google scholar
[9]
Ou Y, Cao S, Zhang Y, et al. Bioprinting microporous functional living materials from protein-based core-shell microgels. Nat Commun. 2023;14:322.
CrossRef Google scholar
[10]
Bankó P, Lee SY, Nagygyörgy V, et al. Technologies for circulating tumor cell separation from whole blood. J Hematol Oncol. 2019;12:48.
CrossRef Google scholar
[11]
Poudineh M, Sargent EH, PantelS K, Kelley O. Profiling circulating tumour cells and other biomarkers of invasive cancers. Nat Biomed Eng. 2018;2:72-84.
CrossRef Google scholar
[12]
Chen S, Cheng J, Kong C-W. et al. Laser-induced fusion of human embryonic stem cells with optical tweezers. Appl Phys Lett. 2013;103:033701.
CrossRef Google scholar
[13]
Yan HHN, Siu HC, Law S, et al. A comprehensive human gastric cancer organoid biobank captures tumor subtype heterogeneity and enables therapeutic screening. Cell Stem Cell. 2018;23:882-897.e11.
CrossRef Google scholar
[14]
Lee W, Tseng P, Di Carlo D. Microtechnology for Cell Manipulation and Sorting. Springer;2017.
[15]
Murphy SV, Atala A. 3D bioprinting of tissues and organs. Nat Biotechnol. 2014;32:773-785.
CrossRef Google scholar
[16]
Turcan I, Olariu MA. Dielectrophoretic manipulation of cancer cells and their electrical characterization. ACS Comb Sci. 2020;22:554-578.
CrossRef Google scholar
[17]
Voldman J. Electrical forces for microscale cell manipulation. Annu Rev Biomed Eng. 2006;8:425-454.
CrossRef Google scholar
[18]
Farahinia A, Zhang WJ, Badea I. Novel microfluidic approaches to circulating tumor cell separation and sorting of blood cells: a review. J Sci: Adv Mater Devices. 2021;6:303-320.
CrossRef Google scholar
[19]
Heidrich H-G, Hannig K. Separation of cell populations by free-flow electrophoresis. Methods in Enzymology. Elsevier;1989.
[20]
Sohail A, Jiang X, Wahid A, et al. Free-flow zone electrophoresis facilitated proteomics analysis of heterogeneous subpopulations in H1299 lung cancer cells. Anal Chim Acta. 2022;1227:340306.
CrossRef Google scholar
[21]
Tomlinson MJ, Tomlinson S, Yang XB, Kirkham J. Cell separation: terminology and practical considerations. J Tissue Eng. 2012;4:2041731412472690.
CrossRef Google scholar
[22]
Lee Y, Kwon J-S. Microfluidic free-flow electrophoresis: a promising tool for protein purification and analysis in proteomics. J Ind Eng Chem. 2022;109:79-99.
CrossRef Google scholar
[23]
Isozaki A, Nakagawa Y, Loo MH, et al. Sequentially addressable dielectrophoretic array for high-throughput sorting of large-volume biological compartments. Sci Adv. 2020;6:eaba6712.
[24]
Gao W, Zhang C, Cai Y, et al. Dielectrophoretic cell sorting with high velocity enabled by two-layer sidewall microelectrodes extending along the entire channel. Sens Actuators B Chem. 2024;410:135669.
CrossRef Google scholar
[25]
Jiang X, Liu S, Zhang Y, et al. Free-flow isoelectric focusing for comprehensive separation and analysis of human salivary microbiome for lung cancer. Anal Chem. 2020;92:12017-12025.
CrossRef Google scholar
[26]
Agarwal M, Desai P, Padh H. Isolation and characterization of subcellular organelles from plant cells. In: WV Dashek, GS Miglani, eds. Plant Cells and Their Organelles. Wiley;2017:25-41.
CrossRef Google scholar
[27]
Basu S, Campbell HM, Dittel BN, Ray A. Purification of specific cell population by fluorescence activated cell sorting (FACS). J Vis Exp. 2010;10:1546.
[28]
Mayer G, Ahmed M-SL. Dolf A, et al. Fluorescence-activated cell sorting for aptamer SELEX with cell mixtures. Nat Protoc. 2010;5:1993-2004.
CrossRef Google scholar
[29]
Bonner W, Hulett H, Sweet R, Herzenberg L. Fluorescence activated cell sorting. Rev Sci Instrum. 1972;43:404-409.
CrossRef Google scholar
[30]
Telford WG. Flow cytometry and cell sorting. Front Med. 2023;10:1287884.
CrossRef Google scholar
[31]
Guo F, Ji X-H. Liu K, et al. Droplet electric separator microfluidic device for cell sorting. Appl Phys Lett. 2010;96:193701.
CrossRef Google scholar
[32]
Fong CY, Peh GSL, Gauthaman K, Bongso A. Separation of SSEA-4 and TRA-1-60 labelled undifferentiated human embryonic stem cells from a heterogeneous cell population using magnetic-activated cell sorting (MACS) and fluorescence-activated cell sorting (FACS). Stem Cell Rev Rep. 2009;5:72-80.
CrossRef Google scholar
[33]
Staubach S, Tertel T, Walkenfort B, et al. Free flow electrophoresis allows quick and reproducible preparation of extracellular vesicles from conditioned cell culture media. Extracellular Vesicles Circulating Nucleic Acids. 2022;3:31-48.
[34]
Liu S, Madren S, Feng P, Sosic Z. Characterization of the acidic species of a monoclonal antibody using free flow electrophoresis fractionation and mass spectrometry. J Pharm Biomed Anal. 2020;185:113217.
CrossRef Google scholar
[35]
Poggel M, Melin T. Free-flow zone electrophoresis: a novel approach and scale-up for preparative protein separation. Electrophoresis. 2001;22:1008-1015.
CrossRef Google scholar
[36]
Courtney M, Thompson E, Glawdel T, Ren CL. Counterflow gradient focusing in free-flow electrophoresis for protein fractionation. Anal Chem. 2020;92:7317-7324.
CrossRef Google scholar
[37]
Lu N, Tay HM, Petchakup C, et al. Label-free microfluidic cell sorting and detection for rapid blood analysis. Lab Chip. 2023;23:1226-1257.
CrossRef Google scholar
[38]
Velmanickam L, Jayasooriya V, Vemuri MS, Tida UR, Nawarathna D. Recent advances in dielectrophoresis toward biomarker detection: a summary of studies published between 2014 and 2021. Electrophoresis. 2022;43:212-231.
CrossRef Google scholar
[39]
Pethig R, Markx GH. Applications of dielectrophoresis in biotechnology. Trends Biotechnol. 1997;15:426-432.
CrossRef Google scholar
[40]
Wang Q, Jones A-AD. Gralnick JA, Lin L, Buie CR. Microfluidic dielectrophoresis illuminates the relationship between microbial cell envelope polarizability and electrochemical activity. Sci Adv. 2019;5:eaat5664.
CrossRef Google scholar
[41]
Cheng J, Sheldon EL, Wu L, et al. Preparation and hybridization analysis of DNA/RNA from E. coli on microfabricated bioelectronic chips. Nat Biotechnol. 1998;16:541-546.
CrossRef Google scholar
[42]
Yu E-S, Lee H, Lee S-M. et al. Precise capture and dynamic relocation of nanoparticulate biomolecules through dielectrophoretic enhancement by vertical nanogap architectures. Nat Commun. 2020;11:2804.
CrossRef Google scholar
[43]
Huang J-Y, Xia L, Xiao X-H. Li G-K. Advances in microchip electrophoresis for the separation and analysis of biological samples. Chin J Chromatogr. 2023;41:641-650.
CrossRef Google scholar
[44]
Li Y, Wang Y, Wan K, et al. On the design, functions, and biomedical applications of high-throughput dielectrophoretic micro-/nanoplatforms: a review. Nanoscale. 2021;13:4330-4358.
CrossRef Google scholar
[45]
Meng X, Sun P, Xu H, Wang Z. Folic acid-functionalized magnetic nanoprobes via a PAMAM dendrimer/SA-biotin mediated cascade-amplifying system for the efficient enrichment of circulating tumor cells. Biomater Sci. 2020;8:6395-6403.
CrossRef Google scholar
[46]
Li F, Xu H, Zhao Y. Magnetic particles as promising circulating tumor cell catchers assisting liquid biopsy in cancer diagnosis: a review. Trends Anal Chem. 2021;145:116453.
CrossRef Google scholar
[47]
Zborowski M, Chalmers JJ. Rare cell separation and analysis by magnetic sorting. Anal Chem. 2011;83:8050-8056.
CrossRef Google scholar
[48]
Borlido L, Moura L, Azevedo AM, et al. Stimuli-responsive magnetic nanoparticles for monoclonal antibody purification. Biotechnol J. 2013;8:709-717.
CrossRef Google scholar
[49]
Gao J, Li Z, Russell T, Li Z. Antibody affinity purification using metallic nickel particles. J Chromatogr B. 2012;895-896:89-93.
CrossRef Google scholar
[50]
Borlido L, Azevedo AM, Sousa AG, et al. Fishing human monoclonal antibodies from a CHO cell supernatant with boronic acid magnetic particles. J Chromatogr B. 2012;903:163-170.
CrossRef Google scholar
[51]
Qian H, Li C, Zhang Y, Lin Z. Efficient isolation of immunoglobulin G by paramagnetic polymer beads modified with 2-mercapto-4-mythyl-pyrimidine. J Immunol Methods. 2009;343:119-129.
CrossRef Google scholar
[52]
Catherine CB, Adam SGC. Functionalisation of magnetic nanoparticles for applications in biomedicine. J Phys D: Appl Phys. 2003;36: R198.
CrossRef Google scholar
[53]
McCloskey KE, Chalmers JJ, Zborowski M. Magnetic cell separation: characterization of magnetophoretic mobility. Anal Chem. 2003;75:6868-6874.
CrossRef Google scholar
[54]
Chalmers JJ, Xiong Y, Jin X, et al. Quantification of non-specific binding of magnetic micro-and nanoparticles using cell tracking velocimetry: implication for magnetic cell separation and detection. Biotechnol Bioeng. 2010;105:1078-1093.
CrossRef Google scholar
[55]
Gordon R, Hogan CE, Neal ML, et al. A simple magnetic separation method for high-yield isolation of pure primary microglia. J Neurosci Methods. 2011;194:287-296.
CrossRef Google scholar
[56]
Frenea-Robin M, Marchalot J. Basic principles and recent advances in magnetic cell separation. Magnetochemistry. 2022;8:11.
CrossRef Google scholar
[57]
Chen L, Wu L-L. Zhang Z-L, et al. Biofunctionalized magnetic nanospheres-based cell sorting strategy for efficient isolation, detection and subtype analyses of heterogeneous circulating hepatocellular carcinoma cells. Biosens Bioelectron. 2016;85:633-640.
CrossRef Google scholar
[58]
Hoshino K, Huang Y-Y. Lane N, et al. Microchip-based immunomagnetic detection of circulating tumor cells. Lab Chip. 2011;11:3449-3457.
CrossRef Google scholar
[59]
Chen K, Duong BTV, Ahmed SU, et al. A magneto-activated nanoscale cytometry platform for molecular profiling of small extracellular vesicles. Nat Commun. 2023;14:5576.
CrossRef Google scholar
[60]
Karabacak NM, Spuhler PS, Fachin F, et al. Microfluidic, marker-free isolation of circulating tumor cells from blood samples. Nat Protoc. 2014;9:694-710.
CrossRef Google scholar
[61]
Wang Z, Wang H, Lin S, et al. Nanoparticle amplification labeling for high-performance magnetic cell sorting. Nano Lett. 2022;22:4774-4783.
CrossRef Google scholar
[62]
Pamme N. Continuous flow separations in microfluidic devices. Lab Chip. 2007;7:1644-1659.
CrossRef Google scholar
[63]
Pamme N, Manz A. On-chip free-flow magnetophoresis: continuous flow separation of magnetic particles and agglomerates. Anal Chem. 2004;76:7250-7256.
CrossRef Google scholar
[64]
Sun Y, Li H, Cui G, et al. A magnetic nanoparticle assisted microfluidic system for low abundance cell sorting with high recovery. Micro Nano Eng. 2022;15:100136.
CrossRef Google scholar
[65]
Nasiri R, Shamloo A, Akbari J. Design of a hybrid inertial and magnetophoretic microfluidic device for CTCs separation from blood. Micromachines. 2021;12:877.
CrossRef Google scholar
[66]
Chen C-L, Chen K-C. Pan Y-C, et al. Separation and detection of rare cells in a microfluidic disk via negative selection. Lab Chip. 2011;11:474-483.
CrossRef Google scholar
[67]
Gourikutty SBN, Chang C-P. Puiu PD. Microfluidic immunomagnetic cell separation from whole blood. J Chromatogr B Analyt Technol Biomed Life Sci. 2016;1011:77-88.
CrossRef Google scholar
[68]
Steinbrueck P, Yu Z, Bissa U, Brieden J, Dose C. REAlease™ immunomagnetic separation technology with reversible labeling for positive selection of leukocytes. J Immunol. 2018;200:174.8.
CrossRef Google scholar
[69]
Jing Y, Moore LR, Schneider T, et al. Negative selection of hematopoietic progenitor cells by continuous magnetophoresis. Exp Hematol. 2007;35:662-672.
CrossRef Google scholar
[70]
Lara O, Tong X, Zborowski M, Chalmers JJ. Enrichment of rare cancer cells through depletion of normal cells using density and flow-through, immunomagnetic cell separation. Exp Hematol. 2004;32:891-904.
CrossRef Google scholar
[71]
Zhao W, Cheng R, Lim SH, et al. Biocompatible and label-free separation of cancer cells from cell culture lines from white blood cells in ferrofluids. Lab Chip. 2017;17:2243-2255.
CrossRef Google scholar
[72]
Zhang L, Xu Z, Kang Y, Xue P. Three-dimensional microfluidic chip with twin-layer herringbone structure for high efficient tumor cell capture and release via antibody-conjugated magnetic microbeads. Electrophoresis. 2018;39:1452-1459.
CrossRef Google scholar
[73]
Aldridge PM, Mukhopadhyay M, Ahmed SU, et al. Prismatic deflection of live tumor cells and cell clusters. ACS Nano. 2018;12:12692-12700.
CrossRef Google scholar
[74]
Philpott DN, Chen K, Atwal RS, et al. Ultrathroughput immunomagnetic cell sorting platform. Lab Chip. 2022;22:4822-4830.
CrossRef Google scholar
[75]
Garg N, Westerhof TM, Liu V, et al. Whole-blood sorting, enrichment and in situ immunolabeling of cellular subsets using acoustic microstreaming. Microsyst Nanoeng. 2018;4:17085.
CrossRef Google scholar
[76]
Xin Z, Lin G, Lei H, Lue TF, Guo Y. Clinical applications of low-intensity pulsed ultrasound and its potential role in urology. Transl Androl Urol. 2016;5:255-266.
CrossRef Google scholar
[77]
Claes L, Willie B. The enhancement of bone regeneration by ultrasound. Prog Biophys Mol Biol. 2007;93:384-398.
CrossRef Google scholar
[78]
Wang Z, Zhe J. Recent advances in particle and droplet manipulation for lab-on-a-chip devices based on surface acoustic waves. Lab Chip. 2011;11:1280-1285.
CrossRef Google scholar
[79]
Leong T, Johansson L, Juliano P, McArthur SL, Manasseh R. Ultrasonic separation of particulate fluids in small and large scale systems: a review. Ind Eng Chem Res. 2013;52:16555-16576.
CrossRef Google scholar
[80]
Rufo J, Cai F, Friend J, Wiklund M, Huang TJ. Acoustofluidics for biomedical applications. Nat Rev Methods Primers. 2022;2:29.
CrossRef Google scholar
[81]
Ung WL, Mutafopulos K, Spink P, et al. Enhanced surface acoustic wave cell sorting by 3D microfluidic-chip design. Lab Chip. 2017;17:4059-4069.
CrossRef Google scholar
[82]
Marx V. Biophysics: using sound to move cells. Nat Methods. 2015;12:41-44.
CrossRef Google scholar
[83]
Schmid L, Weitz DA, Franke T. Sorting drops and cells with acoustics: acoustic microfluidic fluorescence-activated cell sorter. Lab Chip. 2014;14:3710-3718.
CrossRef Google scholar
[84]
Inui T, Mei J, Imashiro C, et al. Focused surface acoustic wave locally removes cells from culture surface. Lab Chip. 2021;21:1299-1306.
CrossRef Google scholar
[85]
Ding X, Lin S-CS. Kiraly B, et al. On-chip manipulation of single microparticles, cells, and organisms using surface acoustic waves. Proc Natl Acad Sci U S A. 2012;109:11105-11109.
CrossRef Google scholar
[86]
Leibacher I, Reichert P, Dual J. Microfluidic droplet handling by bulk acoustic wave (BAW) acoustophoresis. Lab Chip. 2015;15:2896-2905.
CrossRef Google scholar
[87]
Baudoin M, Thomas J-L. Sahely RA, et al. Spatially selective manipulation of cells with single-beam acoustical tweezers. Nat Commun. 2020;11:4244.
CrossRef Google scholar
[88]
Li P, Liang M, Lu X, et al. Sheathless acoustic fluorescence activated cell sorting (aFACS) with high cell viability. Anal Chem. 2019;91:15425-15435.
CrossRef Google scholar
[89]
Huang P-H, Nama N, Mao Z, et al. A reliable and programmable acoustofluidic pump powered by oscillating sharp-edge structures. Lab Chip. 2014;14:4319-4323.
CrossRef Google scholar
[90]
Ahmed D, Ozcelik A, Bojanala N, et al. Rotational manipulation of single cells and organisms using acoustic waves. Nat Commun. 2016;7:11085.
CrossRef Google scholar
[91]
Yang Y, Dejous C, Hallil H. Trends and applications of surface and bulk acoustic wave devices: a review. Micromachines. 2023;14:43.
CrossRef Google scholar
[92]
Yiannacou K, Sariola V. Controlled manipulation and active sorting of particles inside microfluidic chips using bulk acoustic waves and machine learning. Langmuir. 2021;37:4192-4199.
CrossRef Google scholar
[93]
Meng L, Cai F, Li F, et al. Acoustic tweezers. J Phys D: Appl Phys. 2019;52:273001.
CrossRef Google scholar
[94]
Li Y, Lee C, Chen R, Zhou Q, Shung KK. A feasibility study of in vivo applications of single beam acoustic tweezers. Appl Phys Lett. 2014;105:173701.
CrossRef Google scholar
[95]
Li Y, Hwang JY, Shung KK, Lee J. Single-beam acoustic tweezers: a new tool for microparticle manipulation. Acoust Today. 2013;9:10-13.
CrossRef Google scholar
[96]
Lee J, Teh S-Y. Lee A, et al. Single beam acoustic trapping. Appl Phys Lett. 2009;95:073701.
CrossRef Google scholar
[97]
Gong Z, Baudoin M. Single beam acoustical tweezers based on focused beams: a numerical analysis of two-dimensional and three-dimensional trapping capabilities. Phys Rev Appl. 2022;18:044033.
CrossRef Google scholar
[98]
Grier DG. A revolution in optical manipulation. Nature. 2003;424:810-816.
CrossRef Google scholar
[99]
Lenton IC, Armstrong DJ, Stilgoe AB, Nieminen TA, Rubinsztein-Dunlop H. Orientation of swimming cells with annular beam optical tweezers. Opt Commun. 2020;459:124864.
CrossRef Google scholar
[100]
Hu S, Ye J-Y. Zhao Y, Zhu C-L. Advanced optical tweezers on cell manipulation and analysis. Eur Phys J Plus. 2022;137:1024.
CrossRef Google scholar
[101]
Neuman KC, Block SM. Optical trapping. Rev Sci Instrum. 2004;75:2787-2809.
CrossRef Google scholar
[102]
Ashkin A, Dziedzic JM, Yamane T. Optical trapping and manipulation of single cells using infrared laser beams. Nature. 1987;330:769-771.
CrossRef Google scholar
[103]
Chiou PY, Ohta AT, Wu MC. Massively parallel manipulation of single cells and microparticles using optical images. Nature. 2005;436:370-372.
CrossRef Google scholar
[104]
Xie M. Autonomous robot-aided optical tweezer system for biological cell manipulation. Int J Adv Manuf Technol. 2019;105:4953-4966.
CrossRef Google scholar
[105]
Bustamante CJ, Chemla YR, Liu S, Wang MD. Optical tweezers in single-molecule biophysics. Nat Rev Methods Primers. 2021;1:25.
CrossRef Google scholar
[106]
Shabestari MH, Meijering A, Roos W, Wuite G, Peterman E. Recent advances in biological single-molecule applications of optical tweezers and fluorescence microscopy. Methods Enzymol. 2017;582:85-119.
CrossRef Google scholar
[107]
Capitanio M, Pavone FS. Interrogating biology with force: single molecule high-resolution measurements with optical tweezers. Biophys J. 2013;105:1293-1303.
CrossRef Google scholar
[108]
Xie M, Shakoor A, Wu C. Manipulation of biological cells using a robot-aided optical tweezers system. Micromachines. 2018;9:245.
CrossRef Google scholar
[109]
Li X, Cheah CC, Hu S, Sun D. Dynamic trapping and manipulation of biological cells with optical tweezers. Automatica. 2013;49:1614-1625.
CrossRef Google scholar
[110]
Blazquez-Castro A. Optical tweezers: phototoxicity and thermal stress in cells and biomolecules. Micromachines. 2019;10:507.
CrossRef Google scholar
[111]
Aabo T, Perch-Nielsen IR, Dam JS, et al. Effect of long-and short-term exposure to laser light at 1070 nm on growth of Saccharomyces cerevisiae. J Biomed Opt. 2010;15:041505-041507.
CrossRef Google scholar
[112]
Peterman EJ, Gittes F, Schmidt CF. Laser-induced heating in optical traps. Biophys J. 2003;84:1308-1316.
CrossRef Google scholar
[113]
Ju T, Liu S, Yang J, Sun D. Rapidly exploring random tree algorithm-based path planning for robot-aided optical manipulation of biological cells. IEEE Trans Autom Sci Eng. 2013;11:649-657.
CrossRef Google scholar
[114]
Wang X, Chen S, Kong M, et al. Enhanced cell sorting and manipulation with combined optical tweezer and microfluidic chip technologies. Lab Chip. 2011;11:3656-3662.
CrossRef Google scholar
[115]
Li J, Chen Z, Liu Y, et al. Opto-refrigerative tweezers. Sci Adv. 2021;7:eabh1101.
CrossRef Google scholar
[116]
Sun C-K, Huang Y-C. Cheng PC, Liu H-C. Lin B-L. Cell manipulation by use of diamond microparticles as handles of optical tweezers. J Opt Soc Am B. 2001;18:1483-1489.
CrossRef Google scholar
[117]
Kim SB, Yoon SY, Sung HJ, Kim SS. Cross-type optical particle separation in a microchannel. Anal Chem. 2008;80:2628-2630.
CrossRef Google scholar
[118]
Chowdhury S, Thakur A, Åvec P, et al. Automated manipulation of biological cells using gripper formations controlled by optical tweezers. IEEE Trans Autom Sci Eng. 2013;11:338-347.
CrossRef Google scholar
[119]
Guillotin B, Guillemot F. Cell patterning technologies for organotypic tissue fabrication. Trends Biotechnol. 2011;29:183-190.
CrossRef Google scholar
[120]
Lee J, Sutani A, Kaneko R, et al. In vitro generation of functional murine heart organoids via FGF4 and extracellular matrix. Nat Commun. 2020;11:4283.
CrossRef Google scholar
[121]
Song M, Choi DB, Im JS, et al. Modeling acute myocardial infarction and cardiac fibrosis using human induced pluripotent stem cell-derived multi-cellular heart organoids. Cell Death Dis. 2024;15:308.
CrossRef Google scholar
[122]
Volmert B, Kiselev A, Juhong A, et al. A patterned human primitive heart organoid model generated by pluripotent stem cell self-organization. Nat Commun. 2023;14:8245.
CrossRef Google scholar
[123]
Fernandes I, Funakoshi S, Hamidzada H, Epelman S, Keller G. Modeling cardiac fibroblast heterogeneity from human pluripotent stem cell-derived epicardial cells. Nat Commun. 2023;14:8183.
CrossRef Google scholar
[124]
Sarieva K, Kagermeier T, Khakipoor S, et al. Human brain organoid model of maternal immune activation identifies radial glia cells as selectively vulnerable. Mol Psychiatry. 2023;28:5077-5089.
CrossRef Google scholar
[125]
Park DS, Kozaki T, Tiwari SK, et al. iPS-cell-derived microglia promote brain organoid maturation via cholesterol transfer. Nature. 2023;623:397-405.
CrossRef Google scholar
[126]
Jeong GS, No DY, Lee J, et al. Viscoelastic lithography for fabricating self-organizing soft micro-honeycomb structures with ultra-high aspect ratios. Nat Commun. 2016;7:11269.
CrossRef Google scholar
[127]
Jiang S, Xu F, Jin M, et al. Development of a high-throughput micropatterned agarose scaffold for consistent and reproducible hPSC-derived liver organoids. Biofabrication. 2023;15:015006.
CrossRef Google scholar
[128]
Torizal FG, Utami T, Lau QY, et al. Dialysis based-culture medium conditioning improved the generation of human induced pluripotent stem cell derived-liver organoid in a high cell density. Sci Rep. 2022;12:20774.
CrossRef Google scholar
[129]
Vanslambrouck JM, Tan KS, Mah S, Little MH. Generation of proximal tubule-enhanced kidney organoids from human pluripotent stem cells. Nat Protoc. 2023;18:3229-3252.
CrossRef Google scholar
[130]
Lassé M, El Saghir J, Berthier CC, et al. An integrated organoid omics map extends modeling potential of kidney disease. Nat Commun. 2023;14:4903.
CrossRef Google scholar
[131]
Freedman BS, Brooks CR, Lam AQ, et al. Modelling kidney disease with CRISPR-mutant kidney organoids derived from human pluripotent epiblast spheroids. Nat Commun. 2015;6:8715.
CrossRef Google scholar
[132]
Huang L, Holtzinger A, Jagan I, et al. Ductal pancreatic cancer modeling and drug screening using human pluripotent stem cell-and patient-derived tumor organoids. Nat Med. 2015;21:1364-1371.
CrossRef Google scholar
[133]
Lindemans CA, Calafiore M, Mertelsmann AM, et al. Interleukin-22 promotes intestinal-stem-cell-mediated epithelial regeneration. Nature. 2015;528:560-564.
CrossRef Google scholar
[134]
Hahn S, Nam M-O. Noh JH, et al. Organoid-based epithelial to mesenchymal transition (OEMT) model: from an intestinal fibrosis perspective. Sci Rep. 2017;7:2435.
CrossRef Google scholar
[135]
Liu T, Zhou C, Shao Y, et al. Construction and application of in vitro alveolar models based on 3D printing technology. Chin J Mech Eng: Additive Manuf Front. 2022;1:100025.
CrossRef Google scholar
[136]
Kim M, Mun H, Sung CO, et al. Patient-derived lung cancer organoids as in vitro cancer models for therapeutic screening. Nat Commun. 2019;10:3991.
CrossRef Google scholar
[137]
Trisno SL, Philo KED, McCracken KW, et al. Esophageal organoids from human pluripotent stem cells delineate Sox2 functions during esophageal specification. Cell Stem Cell. 2018;23:501-515.e7.
CrossRef Google scholar
[138]
Frenz-Wiessner S, Fairley SD, Buser M, et al. Generation of complex bone marrow organoids from human induced pluripotent stem cells. Nat Methods. 2024;21:868-881.
CrossRef Google scholar
[139]
Limraksasin P, Nattasit P, Manokawinchoke J, et al. Application of shear stress for enhanced osteogenic differentiation of mouse induced pluripotent stem cells. Sci Rep. 2022;12:19021.
CrossRef Google scholar
[140]
Lee J, Rabbani CC, Gao H, et al. Hair-bearing human skin generated entirely from pluripotent stem cells. Nature. 2020;582:399-404.
CrossRef Google scholar
[141]
Lee J, van der Valk WH, Serdy SA, et al. Generation and characterization of hair-bearing skin organoids from human pluripotent stem cells. Nat Protoc. 2022;17:1266-1305.
CrossRef Google scholar
[142]
DiStefano T, Chen HY, Panebianco C, et al. Accelerated and improved differentiation of retinal organoids from pluripotent stem cells in rotating-wall vessel bioreactors. Stem Cell Rep. 2018;10:300-313.
CrossRef Google scholar
[143]
Ramos SA, Armitage LH, Morton JJ, et al. Generation of functional thymic organoids from human pluripotent stem cells. Stem Cell Rep. 2023;18:829-840.
CrossRef Google scholar
[144]
Lomeli-Martin A, Ahamed N, Abhyankar VV, Lapizco-Encinas BH. Electropatterning-contemporary developments for selective particle arrangements employing electrokinetics. Electrophoresis. 2023;44:884-909.
CrossRef Google scholar
[145]
Henslee EA, Dunlop CM, de Mel CM, et al. DEP-Dots for 3D cell culture: low-cost, high-repeatability, effective 3D cell culture in multiple gel systems. Sci Rep. 2020;10:14603.
CrossRef Google scholar
[146]
Albrecht DR, Underhill GH, Mendelson A, Bhatia SN. Multiphase electropatterning of cells and biomaterials. Lab Chip. 2007;7:702-709.
CrossRef Google scholar
[147]
Albrecht DR, Underhill GH, Wassermann TB, Sah RL, Bhatia SN. Probing the role of multicellular organization in three-dimensional microenvironments. Nat Methods. 2006;3:369-375.
CrossRef Google scholar
[148]
Goel M, Verma A, Gupta S. Electric-field driven assembly of live bacterial cell microarrays for rapid phenotypic assessment and cell viability testing. Biosens Bioelectron. 2018;111:159-165.
CrossRef Google scholar
[149]
Albrecht DR, Tsang VL, Sah RL, Bhatia SN. Photo-and electropatterning of hydrogel-encapsulated living cell arrays. Lab Chip. 2005;5:111-118.
CrossRef Google scholar
[150]
Iliescu C, Xu G, Tong WH, et al. Cell patterning using a dielectrophoretic-hydrodynamic trap. Microfluid Nanofluid. 2015;19:363-373.
CrossRef Google scholar
[151]
Ho C-T, Lin R-Z. Chang WY, Chang W-Y. Liu C-H. Rapid heterogeneous liver-cell on-chip patterning via the enhanced field-induced dielectrophoresis trap. Lab Chip. 2006;6:724-734.
CrossRef Google scholar
[152]
Saliba AE, Saias L, Psychari E, et al. Microfluidic sorting and multimodal typing of cancer cells in self-assembled magnetic arrays. Proc Natl Acad Sci U S A. 2010;107:14524-14529.
CrossRef Google scholar
[153]
Gao Q-H, Zhang W-M. Zou H-X, et al. Label-free manipulation via the magneto-Archimedes effect: fundamentals, methodology and applications. Mater Horizons. 2019;6:1359-1379.
CrossRef Google scholar
[154]
Akiyama Y, Morishima K. Label-free cell aggregate formation based on the magneto-Archimedes effect. Appl Phys Lett. 2011;98:163702.
CrossRef Google scholar
[155]
Parfenov VA, Khesuani YD, Petrov SV, et al. Magnetic levitational bioassembly of 3D tissue construct in space. Sci Adv. 2020;6:eaba4174.
CrossRef Google scholar
[156]
Melde K, Athanassiadis AG, Missirlis D, et al. Ultrasound-assisted tissue engineering. Nat Rev Bioeng. 2024;2:486-500.
CrossRef Google scholar
[157]
Kang B, Shin J, Park HJ, et al. High-resolution acoustophoretic 3D cell patterning to construct functional collateral cylindroids for ischemia therapy. Nat Commun. 2018;9:5402.
CrossRef Google scholar
[158]
Comeau ES, Hocking DC, Dalecki D. Ultrasound patterning technologies for studying vascular morphogenesis in 3D. J Cell Sci. 2017;130:232-242.
[159]
Bouyer C, Chen P, Guven S, et al. A bio-acoustic levitational (BAL) assembly method for engineering of multilayered, 3D brain-like constructs, using human embryonic stem cell derived neuro-progenitors. Adv Mater. 2016;28:161-167.
CrossRef Google scholar
[160]
Gu L, Jiang S, Xu X, et al. Size-and density-dependent acoustic differential bioassembly of spatially-defined heterocellular architecture. Biofabrication. 2022;15:015019.
CrossRef Google scholar
[161]
Chen P, Luo Z, Güven S, et al. Microscale assembly directed by liquid-based template. Adv Mater. 2014;26:5936-5941.
CrossRef Google scholar
[162]
Armstrong JPK, Puetzer JL, Serio A, et al. Engineering anisotropic muscle tissue using acoustic cell patterning. Adv Mater. 2018;30:e1802649.
CrossRef Google scholar
[163]
Armstrong JPK, Pchelintseva E, Treumuth S, et al. Tissue engineering cartilage with deep zone cytoarchitecture by high-resolution acoustic cell patterning. Adv Healthc Mater. 2022;11:e2200481.
CrossRef Google scholar
[164]
Wang J, Qiao H, Wang Z, et al. Rational design and acoustic assembly of human cerebral cortex-like microtissues from hiPSC-derived neural progenitors and neurons. Adv Mater. 2023;35:e2210631.
CrossRef Google scholar
[165]
Ren T, Steiger W, Chen P, Ovsianikov A, Demirci U. Enhancing cell packing in buckyballs by acoustofluidic activation. Biofabrication. 2020;12:025033.
CrossRef Google scholar
[166]
Ren T, Chen P, Gu L, Ogut MG, Demirci U. Soft ring-shaped cellu-robots with simultaneous locomotion in batches. Adv Mater. 2020;32:e1905713.
CrossRef Google scholar
[167]
Chen P, Guven S, Usta OB, Yarmush ML, Demirci U. Biotunable acoustic node assembly of organoids. Adv Healthc Mater. 2015;4:1937-1943.
CrossRef Google scholar
[168]
Gu Y, Chen C, Rufo J, et al. Acoustofluidic holography for micro-to nanoscale particle manipulation. ACS Nano. 2020;14:14635-14645.
CrossRef Google scholar
[169]
Ozcelik A, Rufo J, Guo F, et al. Acoustic tweezers for the life sciences. Nat Methods. 2018;15:1021-1028.
CrossRef Google scholar
[170]
Yang S, Tian Z, Wang Z, et al. Harmonic acoustics for dynamic and selective particle manipulation. Nat Mater. 2022;21:540-546.
CrossRef Google scholar
[171]
Yang S, Rufo J, Zhong R, et al. Acoustic tweezers for high-throughput single-cell analysis. Nat Protoc. 2023;18:2441-2458.
CrossRef Google scholar
[172]
Tian Z, Wang Z, Zhang P, et al. Generating multifunctional acoustic tweezers in Petri dishes for contactless, precise manipulation of bioparticles. Sci Adv. 2020;6:eabb0494.
CrossRef Google scholar
[173]
Ma Z, Holle AW, Melde K, et al. Acoustic holographic cell patterning in a biocompatible hydrogel. Adv Mater. 2020;32:e1904181.
CrossRef Google scholar
[174]
Cox L, Melde K, Croxford A, Fischer P, Drinkwater BW. Acoustic hologram enhanced phased arrays for ultrasonic particle manipulation. Phys Rev Appl. 2019;12:064055.
CrossRef Google scholar
[175]
Parfenov VA, Koudan EV, Krokhmal AA, et al. Biofabrication of a functional tubular construct from tissue spheroids using magnetoacoustic levitational directed assembly. Adv Healthc Mater. 2020;9:e2000721.
CrossRef Google scholar
[176]
Li W, Xu Z, Huang J, et al. NeuroArray: a universal interface for patterning and interrogating neural circuitry with single cell resolution. Sci Rep. 2014;4:4784.
CrossRef Google scholar
[177]
Khetani SR, Bhatia SN. Microscale culture of human liver cells for drug development. Nat Biotechnol. 2008;26:120-126.
CrossRef Google scholar
[178]
Lee I, Ludwig AL, Phillips MJ, et al. Ultrathin micromolded 3D scaffolds for high-density photoreceptor layer reconstruction. Sci Adv. 2021;7:eabf0344.
CrossRef Google scholar
[179]
Jung YH, Phillips MJ, Lee J, et al. 3D microstructured scaffolds to support photoreceptor polarization and maturation. Adv Mater. 2018;30:e1803550.
CrossRef Google scholar
[180]
Wiedenmann S, Breunig M, Merkle J, et al. Single-cell-resolved differentiation of human induced pluripotent stem cells into pancreatic duct-like organoids on a microwell chip. Nat Biomed Eng. 2021;5:897-913.
CrossRef Google scholar
[181]
Ma Y, Han X, Castro RB, et al. Analysis of the bystander effect in cone photoreceptors via a guided neural network platform. Sci Adv. 2018;4:eaas9274.
CrossRef Google scholar
[182]
Flaim CJ, Chien S, Bhatia SN. An extracellular matrix microarray for probing cellular differentiation. Nat Methods. 2005;2:119-125.
CrossRef Google scholar
[183]
Zhai J, Li H, Wong AH-H. et al. A digital microfluidic system with 3D microstructures for single-cell culture. Microsyst Nanoeng. 2020;6:6.
CrossRef Google scholar
[184]
Boya M, Ozkaya-Ahmadov T, Swain BE, et al. High throughput, label-free isolation of circulating tumor cell clusters in meshed microwells. Nat Commun. 2022;13:3385.
CrossRef Google scholar
[185]
Li C, Ouyang L, Pence IJ, et al. Buoyancy-driven gradients for biomaterial fabrication and tissue engineering. Adv Mater. 2019;31:e1900291.
CrossRef Google scholar
[186]
Goulart E, de Caires-Junio LC, Telles-Silva KA, et al. 3D bioprinting of liver spheroids derived from human induced pluripotent stem cells sustain liver function and viability in vitro. Biofabrication. 2019;12:015010.
CrossRef Google scholar
[187]
Mahfouzi SH, Safiabadi Tali SH, Amoabediny G. 3D bioprinting for lung and tracheal tissue engineering: criteria, advances, challenges, and future directions. Bioprinting. 2021;21:e00124.
CrossRef Google scholar
[188]
Zhang YS, Haghiashtiani G, Hübscher T, et al. 3D extrusion bioprinting. Nat Rev Methods Primers. 2021;1:75.
CrossRef Google scholar
[189]
Colosi C, Shin SR, Manoharan V, et al. Microfluidic bioprinting of heterogeneous 3D tissue constructs using low-viscosity bioink. Adv Mater. 2016;28:677-684.
CrossRef Google scholar
[190]
Costantini M, Colosi C, Swieszkowski W, Barbetta A. Co-axial wet-spinning in 3D bioprinting: state of the art and future perspective of microfluidic integration. Biofabrication. 2018;11:012001.
CrossRef Google scholar
[191]
Gao G, Park W, Kim BS, et al. Construction of a novel in vitro atherosclerotic model from geometry-tunable artery equivalents engineered via in-bath coaxial cell printing. Adv Funct Mater. 2020;31:2008878.
CrossRef Google scholar
[192]
Jia W, Gungor-Ozkerim PS, Zhang YS, et al. Direct 3D bioprinting of perfusable vascular constructs using a blend bioink. Biomaterials. 2016;106:58-68.
CrossRef Google scholar
[193]
Onoe H, Okitsu T, Itou A, et al. Metre-long cell-laden microfibres exhibit tissue morphologies and functions. Nat Mater. 2013;12:584-590.
CrossRef Google scholar
[194]
Wang D, Maharjan S, Kuang X, et al. Microfluidic bioprinting of tough hydrogel-based vascular conduits for functional blood vessels. Sci Adv. 2022;8:eabq6900.
CrossRef Google scholar
[195]
Kang E, Jeong GS, Choi YY, et al. Digitally tunable physicochemical coding of material composition and topography in continuous microfibres. Nat Mater. 2011;10:877-883.
CrossRef Google scholar
[196]
Ahrens JH, Uzel SGM, Skylar-Scott M, et al. Programming cellular alignment in engineered cardiac tissue via bioprinting anisotropic organ building blocks. Adv Mater. 2022;34:e2200217.
CrossRef Google scholar
[197]
Zhang P, Abate AR. High-definition single-cell printing: cell-by-cell fabrication of biological structures. Adv Mater. 2020;32:e2005346.
CrossRef Google scholar
[198]
Jeffries GDM, Xu S, Lobovkina T, et al. 3D micro-organisation printing of mammalian cells to generate biological tissues. Sci Rep. 2020;10:19529.
CrossRef Google scholar
[199]
Ayan B, Celik N, Zhang Z, et al. Aspiration-assisted freeform bioprinting of prefabricated tissue spheroids in a yield-stress gel. Commun Phys. 2020;3:183.
CrossRef Google scholar
[200]
Cao Y, Tan J, Zhao H, et al. Bead-jet printing enabled sparse mesenchymal stem cell patterning augments skeletal muscle and hair follicle regeneration. Nat Commun. 2022;13:7463.
CrossRef Google scholar
[201]
Zhang Y, Hu Q, Pei Y, et al. A patient-specific lung cancer assembloid model with heterogeneous tumor microenvironments. Nat Commun. 2024;15:3382.
CrossRef Google scholar
[202]
Levato R, Dudaryeva O, Garciamendez-Mijares CE, et al. Light-based vat-polymerization bioprinting. Nat Rev Methods Primers. 2023;3:47.
CrossRef Google scholar
[203]
Li W, Wang M, Ma H, et al. Stereolithography apparatus and digital light processing-based 3D bioprinting for tissue fabrication. iScience. 2023;26:106039.
CrossRef Google scholar
[204]
Zorlutuna P, Jeong JH, Kong H, Bashir R. Stereolithography-based hydrogel microenvironments to examine cellular interactions. Adv Funct Mater. 2011;21:3642-3651.
CrossRef Google scholar
[205]
Grigoryan B, Paulsen SJ, Corbett DC, et al. Multivascular networks and functional intravascular topologies within biocompatible hydrogels. Science. 2019;364:458-464.
CrossRef Google scholar
[206]
Wang Z, Kumar H, Tian Z, et al. Visible light photoinitiation of cell-adhesive gelatin methacryloyl hydrogels for stereolithography 3D bioprinting. ACS Appl Mater Interfaces. 2018;10:26859-26869.
CrossRef Google scholar
[207]
Zhou X, Zhu W, Nowicki M, et al. 3D bioprinting a cell-laden bone matrix for breast cancer metastasis study. ACS Appl Mater Interfaces. 2016;8:30017-30026.
CrossRef Google scholar
[208]
Tang M, Xie Q, Gimple RC, et al. Three-dimensional bioprinted glioblastoma microenvironments model cellular dependencies and immune interactions. Cell Res. 2020;30:833-853.
CrossRef Google scholar
[209]
Madrid-Wolff J, Boniface A, Loterie D, Delrot P, Moser C. Controlling light in scattering materials for volumetric additive manufacturing. Adv Sci. 2022;9:e2105144.
CrossRef Google scholar
[210]
Tang M, Tiwari SK, Agrawal K, et al. Rapid 3D bioprinting of glioblastoma model mimicking native biophysical heterogeneity. Small. 2021;17:e2006050.
CrossRef Google scholar
[211]
Bernal PN, Delrot P, Loterie D, et al. Volumetric bioprinting of complex living-tissue constructs within seconds. Adv Mater. 2019;31:e1904209.
CrossRef Google scholar
[212]
Xu H, Casillas J, Krishnamoorthy S, Xu C. Effects of Irgacure 2959 and lithium phenyl-2, 4, 6-trimethylbenzoylphosphinate on cell viability, physical properties, and microstructure in 3D bioprinting of vascular-like constructs. Biomed Mater. 2020;15:055021.
CrossRef Google scholar

RIGHTS & PERMISSIONS

2025 2025 The Author(s). Droplet published by Jilin University and John Wiley & Sons Australia, Ltd.
PDF

Accesses

Citations

Detail

Sections
Recommended

/