DOI QR코드

DOI QR Code

Monitoring the Differentiation and Migration Patterns of Neural Cells Derived from Human Embryonic Stem Cells Using a Microfluidic Culture System

  • Lee, Nayeon (CHA Stem Cell Institute, CHA University) ;
  • Park, Jae Woo (Division of World Class University Multiscale Mechanical Design, School of Mechanical and Aerospace Engineering, Seoul National University) ;
  • Kim, Hyung Joon (Division of World Class University Multiscale Mechanical Design, School of Mechanical and Aerospace Engineering, Seoul National University) ;
  • Yeon, Ju Hun (Division of World Class University Multiscale Mechanical Design, School of Mechanical and Aerospace Engineering, Seoul National University) ;
  • Kwon, Jihye (CHA Stem Cell Institute, CHA University) ;
  • Ko, Jung Jae (CHA Stem Cell Institute, CHA University) ;
  • Oh, Seung-Hun (CHA Stem Cell Institute, CHA University) ;
  • Kim, Hyun Sook (CHA Stem Cell Institute, CHA University) ;
  • Kim, Aeri (CHA Stem Cell Institute, CHA University) ;
  • Han, Baek Soo (Research Center for Integrated Cellulomics, Korea Research Institute of Bioscience and Biotechnology) ;
  • Lee, Sang Chul (Research Center for Integrated Cellulomics, Korea Research Institute of Bioscience and Biotechnology) ;
  • Jeon, Noo Li (Division of World Class University Multiscale Mechanical Design, School of Mechanical and Aerospace Engineering, Seoul National University) ;
  • Song, Jihwan (CHA Stem Cell Institute, CHA University)
  • Received : 2014.05.26
  • Accepted : 2014.05.29
  • Published : 2014.06.30

Abstract

Microfluidics can provide unique experimental tools to visualize the development of neural structures within a microscale device, which is followed by guidance of neurite growth in the axonal isolation compartment. We utilized microfluidics technology to monitor the differentiation and migration of neural cells derived from human embryonic stem cells (hESCs). We co-cultured hESCs with PA6 stromal cells, and isolated neural rosette-like structures, which subsequently formed neurospheres in suspension culture. Tuj1-positive neural cells, but not nestin-positive neural precursor cells (NPCs), were able to enter the microfluidics grooves (microchannels), suggesting that neural cell-migratory capacity was dependent upon neuronal differentiation stage. We also showed that bundles of axons formed and extended into the microchannels. Taken together, these results demonstrated that microfluidics technology can provide useful tools to study neurite outgrowth and axon guidance of neural cells, which are derived from human embryonic stem cells.

Keywords

References

  1. Bellin, M., Marchetto, M.C., Gage, F.H., and Mummery, C.L. (2012). Induced pluripotent stem cells: the new patient? Nat. Rev. Mol. Cell. Biol. 13, 713-726.
  2. Blow, N. (2008). Stem cells: in search of common ground. Nature 451, 855-858. https://doi.org/10.1038/451855a
  3. Breslauer, D.N., Lee, P.J., and Lee LP. (2006). Microfluidics-based systems biology. Mol. Biosyst. 2, 97-112. https://doi.org/10.1039/b515632g
  4. Cohen, M.S., Bas Orth, C., Kim, H.J., Jeon, N.L., and Jaffrey, S,R. (2011). Neurotrophin-mediated dendrite-to-nucleus signaling revealed by microfluidic compartmentalization of dendrites. Proc. Natl. Acad. Sci. USA 108, 11246-11251. https://doi.org/10.1073/pnas.1012401108
  5. Hamon, M., and Hong, J.W. (2013). New tools and new biology: recent miniaturized systems for molecular and cellular biology. Mol. Cells 36, 485-506. https://doi.org/10.1007/s10059-013-0333-1
  6. Jeon, I., Lee, N., Li, J.Y., Park, I.H., Park, S., Moon, J., Shim, S.H., Choi, C., Chang, D.J., Kwon, J., et al. (2012). Neural properties, in vivo effects and pathology of a Huntington's disease patient-derived induced pluripotent stem cells. Stem Cells 30, 2054-2062. https://doi.org/10.1002/stem.1135
  7. Kim, H.J., Park, J.W., Byun, J.H., Vahidi, B., Rhee, S.W., and Jeon, N.L. (2012). Integrated microfluidics platforms for investigating injury and regeneration of CNS axons. Ann. Biomed. Eng. 40, 1268-1276. https://doi.org/10.1007/s10439-012-0515-6
  8. Kosik, K.S., and Finch, E.A. (1987). MAP2 and Tau segregate into dendrite and axon domains after the elaboration of morphologically distinct neuritis: an immunocytochemical study of cultured rat cerebrum. J. Neurosci. 7, 3142-3153.
  9. Lee, N., Choi, C., Jeon, I., and Song, J. (2009). Differentiation of GABAergic neurons from human embryonic stem cells. Tissue Eng. Regen. Med. 6, 1359-1365.
  10. Millet, L.J., and Gillette, M.U. (2012). New perspectives on neuronal development via microfluidic environments. Trends Neurosci. 35, 752-761. https://doi.org/10.1016/j.tins.2012.09.001
  11. Millet, L.J., Stewart, M.E., Nuzzo, R.G., and Gillette, M.U. (2010). Guiding neuron development with planar surface gradients of substrate cues deposited using microfluidic devices. Lab. Chip 10, 1525-1535. https://doi.org/10.1039/c001552k
  12. Park, J.W., Vahidi, B., Taylor, A.M., Rhee, S.W., and Jeon, N.L. (2006). Microfluidic culture platform for neuroscience research. Nat. Protoc. 1, 2128-2136. https://doi.org/10.1038/nprot.2006.316
  13. Park, J.W., Kim, H.J., Byun, J.H., Ryu, H.R., and Jeon, N.L. (2009). Novel microfluidic platform for culturing neurons: culturing and biochemical analysis of neuronal components. Biotechnol. J. 4, 1573-1577. https://doi.org/10.1002/biot.200900159
  14. Park, J.W., Kim, H.J., Kang, M.W., and Jeon, N.L. (2013). Advances in microfluidics-based experimental methods for neuroscience research. Lab. Chip 13, 509-521. https://doi.org/10.1039/c2lc41081h
  15. Reubinoff, B.E., Pera, M.F., Fong, C.Y., Trounson, A., and Bongso, A. (2000). Embryonic stem cell lines from human blastocysts: somatic differentiation in vitro. Nat. Biotechnol. 18, 399-404. https://doi.org/10.1038/74447
  16. Rhee, S.W., Taylor, A.M., Tu, C.H., Cribbs, D.H., Cotman, C.W., and Jeon, N.L. (2005). Patterned cell culture inside microfluidic devices. Lab. Chip 2005 5, 102-107. https://doi.org/10.1039/b403091e
  17. Shin, H.S., Kim, H.J., Min, S.K., Kim, S.H., Lee, B.M., and Jeon, N.L. (2010). Compartmental culture of embryonic stem cell-derived neurons in microfluidic devices for use in axonal biology. Biotechnol. Lett. 32, 1063-1070. https://doi.org/10.1007/s10529-010-0280-2
  18. Taylor, A.M., Rhee, S.W., and Jeon, N.L. (2006). Microfluidic chambers for cell migration and neuroscience research. Methods Mol. Biol. 321, 167-177.
  19. Thomson, J.A., Itskovitz-Eldor, J., Shapiro, S.S., Waknitz, M.A., Swiergiel, J.J., Marshall, V.S., and Jones, J.M. (1998). Embryonic stem cell lines derived from human blastocysts. Science 282, 1145-1147. https://doi.org/10.1126/science.282.5391.1145

Cited by

  1. Full biological characterization of human pluripotent stem cells will open the door to translational research vol.90, pp.9, 2016, https://doi.org/10.1007/s00204-016-1763-2
  2. Controlling Differentiation of Stem Cells for Developing Personalized Organ-on-Chip Platforms 2017, https://doi.org/10.1002/adhm.201700426
  3. Analytical Techniques in Neuroscience: Recent Advances in Imaging, Separation, and Electrochemical Methods vol.89, pp.1, 2017, https://doi.org/10.1021/acs.analchem.6b04278
  4. Synaptic compartmentalization by micropatterned masking of a surface adhesive cue in cultured neurons vol.92, 2016, https://doi.org/10.1016/j.biomaterials.2016.03.027
  5. Real-time Monitoring of Discrete Synaptic Release Events and Excitatory Potentials within Self-reconstructed Neuromuscular Junctions vol.127, pp.32, 2015, https://doi.org/10.1002/ange.201503801
  6. Depletion of Inositol Polyphosphate 4-Phosphatase II Suppresses Callosal Axon Formation in the Developing Mice vol.39, pp.6, 2016, https://doi.org/10.14348/molcells.2016.0058
  7. Semaphorin 3C Released from a Biocompatible Hydrogel Guides and Promotes Axonal Growth of Rodent and Human Dopaminergic Neurons vol.22, pp.11-12, 2016, https://doi.org/10.1089/ten.tea.2016.0008
  8. Microfluidic systems for stem cell-based neural tissue engineering vol.16, pp.14, 2016, https://doi.org/10.1039/C6LC00489J
  9. The microbiota and autoimmunity: Their role in thyroid autoimmune diseases vol.183, 2017, https://doi.org/10.1016/j.clim.2017.07.001
  10. Advances in microfluidic platforms for analyzing and regulating human pluripotent stem cells vol.34, 2015, https://doi.org/10.1016/j.gde.2015.07.007
  11. Real-time Monitoring of Discrete Synaptic Release Events and Excitatory Potentials within Self-reconstructed Neuromuscular Junctions vol.54, pp.32, 2015, https://doi.org/10.1002/anie.201503801
  12. A Gal-MµS Device to Evaluate Cell Migratory Response to Combined Galvano-Chemotactic Fields vol.7, pp.4, 2017, https://doi.org/10.3390/bios7040054
  13. Retrograde interferon-gamma signaling induces major histocompatibility class I expression in human-induced pluripotent stem cell-derived neurons vol.5, pp.2, 2017, https://doi.org/10.1002/acn3.516
  14. A monitoring system for axonal growth dynamics using micropatterns of permissive and Semaphorin 3F chemorepulsive signals vol.19, pp.2, 2019, https://doi.org/10.1039/C8LC00845K
  15. Application of microfluidic systems for neural differentiation of cells vol.2, pp.4, 2019, https://doi.org/10.33218/prnano2(4).181127.2
  16. Multi-compartment Microfluidic Device Geometry and Covalently Bound Poly-D-Lysine Influence Neuronal Maturation vol.7, pp.None, 2019, https://doi.org/10.3389/fbioe.2019.00084
  17. Microfluidic three-dimensional cell culture of stem cells for high-throughput analysis vol.11, pp.10, 2019, https://doi.org/10.4252/wjsc.v11.i10.803
  18. Optogenetics for neural transplant manipulation and functional analysis vol.527, pp.2, 2014, https://doi.org/10.1016/j.bbrc.2020.01.141
  19. Novel Concept of Micro Patterned Micro Titer Plates Fabricated via UV-NIL for Automated Neuronal Cell Assay Read-Out vol.11, pp.4, 2014, https://doi.org/10.3390/nano11040902
  20. Directional Growth of Human Neuronal Axons in a Microfluidic Device with Nanotopography on Azobenzene‐Based Material vol.8, pp.11, 2014, https://doi.org/10.1002/admi.202100048