Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Protocol
  • Published:

3D mouse embryonic stem cell culture for generating inner ear organoids

Abstract

This protocol describes a culture system in which inner-ear sensory tissue is produced from mouse embryonic stem (ES) cells under chemically defined conditions. This model is amenable to basic and translational investigations into inner ear biology and regeneration. In this protocol, mouse ES cells are aggregated in 96-well plates in medium containing extracellular matrix proteins to promote epithelialization. During the first 14 d, a series of precisely timed protein and small-molecule treatments sequentially induce epithelia that represent the mouse embryonic non-neural ectoderm, preplacodal ectoderm and otic vesicle epithelia. Ultimately, these tissues develop into cysts with a pseudostratified epithelium containing inner ear hair cells and supporting cells after 16–20 d. Concurrently, sensory-like neurons generate synapse-like structures with the derived hair cells. We have designated the stem cell–derived epithelia harboring hair cells, supporting cells and sensory-like neurons as inner ear organoids. This method provides a reproducible and scalable means to generate inner ear sensory tissue in vitro.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Overview of inner ear induction protocol.
Figure 2: Development of the definitive ectoderm epithelium.
Figure 3: Morphology changes after BMP/SB-FGF/LDN treatment.
Figure 4: Aggregate organization on day 8 of differentiation.
Figure 5: Cellular rearrangement between days 8 and 12 of differentiation.
Figure 6: Hair cell induction and organoid types.
Figure 7: Inner ear organoid dissection for electrophysiological analysis.

Similar content being viewed by others

References

  1. Sasai, Y. Cytosystems dynamics in self-organization of tissue architecture. Nature 493, 318–326 (2013).

    Article  CAS  PubMed  Google Scholar 

  2. Sasai, Y., Eiraku, M. & Suga, H. In vitro organogenesis in three dimensions: self-organising stem cells. Development 139, 4111–4121 (2012).

    Article  CAS  PubMed  Google Scholar 

  3. Koehler, K.R., Mikosz, A.M., Molosh, A.I., Patel, D. & Hashino, E. Generation of inner ear sensory epithelia from pluripotent stem cells in 3D culture. Nature 500, 217–221 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Gonzalez-Cordero, A. et al. Photoreceptor precursors derived from three-dimensional embryonic stem cell cultures integrate and mature within adult degenerate retina. Nat. Biotechnol. 31, 741–747 (2013).

    Article  CAS  PubMed  Google Scholar 

  5. Eiraku, M. et al. Self-organizing optic-cup morphogenesis in three-dimensional culture. Nature 472, 51–56 (2011).

    Article  CAS  PubMed  Google Scholar 

  6. Eiraku, M. et al. Self-organized formation of polarized cortical tissues from ESCs and its active manipulation by extrinsic signals. Cell Stem Cell 3, 519–532 (2008).

    Article  CAS  PubMed  Google Scholar 

  7. Eiraku, M. & Sasai, Y. Mouse embryonic stem cell culture for generation of three-dimensional retinal and cortical tissues. Nat. Protoc. 7, 69–79 (2012).

    Article  CAS  Google Scholar 

  8. Nakano, T. et al. Self-formation of optic cups and storable stratified neural retina from human ESCs. Cell Stem Cell 10, 771–785 (2012).

    Article  CAS  PubMed  Google Scholar 

  9. Suga, H. et al. Self-formation of functional adenohypophysis in three-dimensional culture. Nature 480, 57–62 (2011).

    Article  CAS  PubMed  Google Scholar 

  10. Nasu, M. et al. Robust formation and maintenance of continuous stratified cortical neuroepithelium by laminin-containing matrix in mouse ES cell culture. PLoS ONE 7, e53024 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Lancaster, M. et al. Cerebral organoids model human brain development and microcephaly. Nature 501, 373–379 (2013).

    Article  CAS  PubMed  Google Scholar 

  12. Spence, J.R. et al. Directed differentiation of human pluripotent stem cells into intestinal tissue in vitro. Nature 470, 105–109 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. McCracken, K.W., Howell, J.C., Wells, J.M. & Spence, J.R. Generating human intestinal tissue from pluripotent stem cells in vitro. Nat. Protoc. 6, 1920–1928 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Sato, T. et al. Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche. Nature 459, 262–265 (2009).

    Article  CAS  PubMed  Google Scholar 

  15. Huch, M. et al. In vitro expansion of single Lgr5+ liver stem cells induced by Wnt-driven regeneration. Nature 494, 247–250 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Li, L. et al. Location of transient ectodermal progenitor potential in mouse development. Development 140, 4533–4543 (2013).

    Article  CAS  PubMed  Google Scholar 

  17. Cajal, M. et al. Clonal and molecular analysis of the prospective anterior neural boundary in the mouse embryo. Development 139, 423–436 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Beddington, S.P. An autoradiographic analysis of the potency of embryonic ectoderm in the 8th day postimplantation mouse embryo. J. Embryol. Exp. Morphol. 64, 87–104 (1981).

    CAS  PubMed  Google Scholar 

  19. Beddington, R.S. An autoradiographic analysis of tissue potency in different regions of the embryonic ectoderm during gastrulation in the mouse. J. Embryol Exp. Morphol. 69, 265–285 (1982).

    CAS  PubMed  Google Scholar 

  20. Wilson, P.A. & Hemmati-Brivanlou, A. Induction of epidermis and inhibition of neural fate by Bmp-4. Nature 376, 331–333 (1995).

    Article  CAS  PubMed  Google Scholar 

  21. Wilson, P.A., Lagna, G., Suzuki, A. & Hemmati-Brivanlou, A. Concentration-dependent patterning of the Xenopus ectoderm by BMP4 and its signal transducer Smad1. Development 124, 3177–3184 (1997).

    Article  CAS  PubMed  Google Scholar 

  22. Kwon, H.-J., Bhat, N., Sweet, E.M., Cornell, R.A. & Riley, B.B. Identification of early requirements for preplacodal ectoderm and sensory organ development. PLoS Genet. 6, e1001133 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  23. Barth, K.A. et al. Bmp activity establishes a gradient of positional information throughout the entire neural plate. Development 126, 4977–4987 (1999).

    Article  CAS  PubMed  Google Scholar 

  24. Neave, B., Holder, N. & Patient, R. A graded response to BMP-4 spatially coordinates patterning of the mesoderm and ectoderm in the zebrafish. Mech. Dev. 62, 183–195 (1997).

    Article  CAS  PubMed  Google Scholar 

  25. Harvey, N.T. et al. Response to BMP4 signalling during ES cell differentiation defines intermediates of the ectoderm lineage. J. Cell Sci. 123, 1796–1804 (2010).

    Article  CAS  PubMed  Google Scholar 

  26. Pieper, M., Ahrens, K., Rink, E., Peter, A. & Schlosser, G. Differential distribution of competence for panplacodal and neural crest induction to non-neural and neural ectoderm. Development 139, 1175–1187 (2012).

    Article  CAS  PubMed  Google Scholar 

  27. Reichert, S., Randall, R.A. & Hill, C.S. A BMP regulatory network controls ectodermal cell fate decisions at the neural plate border. Development 140, 4435–4444 (2013).

    Article  CAS  PubMed  Google Scholar 

  28. Grocott, T., Tambalo, M. & Streit, A. The peripheral sensory nervous system in the vertebrate head: a gene regulatory perspective. Dev. Biol. 370, 3–23 (2012).

    Article  CAS  PubMed  Google Scholar 

  29. Ahrens, K. & Schlosser, G. Tissues and signals involved in the induction of placodal Six1 expression in Xenopus laevis. Dev. Biol. 288, 40–59 (2005).

    Article  CAS  PubMed  Google Scholar 

  30. Brugmann, S.A., Pandur, P.D., Kenyon, K.L., Pignoni, F. & Moody, S.A. Six1 promotes a placodal fate within the lateral neurogenic ectoderm by functioning as both a transcriptional activator and repressor. Development 131, 5871–5881 (2004).

    Article  CAS  PubMed  Google Scholar 

  31. Litsiou, A., Hanson, S. & Streit, A. A balance of FGF, BMP and WNT signalling positions the future placode territory in the head. Development 132, 4051–4062 (2005).

    Article  CAS  PubMed  Google Scholar 

  32. Schlosser, G. Induction and specification of cranial placodes. Dev. Biol. 294, 303–351 (2006).

    Article  CAS  PubMed  Google Scholar 

  33. Patthey, C. & Gunhaga, L. Specification and regionalisation of the neural plate border. Eur. J. Neurosci. 34, 1516–1528 (2011).

    Article  PubMed  Google Scholar 

  34. Bhattacharyya, S. & Bronner-Fraser, M. Competence, specification and commitment to an olfactory placode fate. Development 135, 4165–4177 (2008).

    Article  CAS  PubMed  Google Scholar 

  35. Baker, C.V., Stark, M.R., Marcelle, C. & Bronner-Fraser, M. Competence, specification and induction of Pax-3 in the trigeminal placode. Development 126, 147–156 (1999).

    Article  CAS  PubMed  Google Scholar 

  36. Ladher, R.K., O'Neill, P. & Begbie, J. From shared lineage to distinct functions: the development of the inner ear and epibranchial placodes. Development 137, 1777–1785 (2010).

    Article  CAS  PubMed  Google Scholar 

  37. Wright, T.J. & Mansour, S.L. Fgf3 and Fgf10 are required for mouse otic placode induction. Development 130, 3379–3390 (2003).

    Article  CAS  PubMed  Google Scholar 

  38. Urness, L.D., Paxton, C.N., Wang, X., Schoenwolf, G.C. & Mansour, S.L. FGF signaling regulates otic placode induction and refinement by controlling both ectodermal target genes and hindbrain Wnt8a. Dev. Biol. 340, 595–604 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Ohyama, T., Mohamed, O.A., Taketo, M.M., Dufort, D. & Groves, A.K. Wnt signals mediate a fate decision between otic placode and epidermis. Development 133, 865–875 (2006).

    Article  CAS  PubMed  Google Scholar 

  40. Riccomagno, M.M., Takada, S. & Epstein, D.J. Wnt-dependent regulation of inner ear morphogenesis is balanced by the opposing and supporting roles of Shh. Genes Dev. 19, 1612–1623 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Freter, S., Muta, Y., Mak, S.-S., Rinkwitz, S. & Ladher, R.K. Progressive restriction of otic fate: the role of FGF and Wnt in resolving inner ear potential. Development 135, 3415–3424 (2008).

    Article  CAS  PubMed  Google Scholar 

  42. Groves, A.K. & Bronner-Fraser, M. Competence, specification and commitment in otic placode induction. Development 127, 3489–3499 (2000).

    Article  CAS  PubMed  Google Scholar 

  43. Groves, A.K. & Fekete, D.M. Shaping sound in space: the regulation of inner ear patterning. Development 139, 245–257 (2011).

    Article  CAS  Google Scholar 

  44. Neves, J., Parada, C., Chamizo, M. & Giraldez, F. Jagged 1 regulates the restriction of Sox2 expression in the developing chicken inner ear: a mechanism for sensory organ specification. Development 138, 735–744 (2011).

    Article  CAS  PubMed  Google Scholar 

  45. Appler, J.M. & Goodrich, L.V. Connecting the ear to the brain: molecular mechanisms of auditory circuit assembly. Prog. Neurobiol. 93, 488–508 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Laine, H., Sulg, M., Kirjavainen, A. & Pirvola, U. Cell cycle regulation in the inner ear sensory epithelia: role of cyclin D1 and cyclin-dependent kinase inhibitors. Dev. Biol. 337, 134–146 (2010).

    Article  CAS  PubMed  Google Scholar 

  47. Oesterle, E.C., Campbell, S., Taylor, R.R., Forge, A. & Hume, C.R. Sox2 and Jagged1 expression in normal and drug-damaged adult mouse inner ear. J. Assoc. Res. Otolaryngol. 9, 65–89 (2007).

    Article  PubMed  PubMed Central  Google Scholar 

  48. Xiang, M., Gao, W.Q., Hasson, T. & Shin, J.J. Requirement for Brn-3c in maturation and survival, but not in fate determination of inner ear hair cells. Development 125, 3935–3946 (1998).

    Article  CAS  PubMed  Google Scholar 

  49. Watanabe, K. et al. Directed differentiation of telencephalic precursors from embryonic stem cells. Nat. Neurosci. 8, 288–296 (2005).

    Article  CAS  PubMed  Google Scholar 

  50. Muñoz-Sanjuán, I. & Brivanlou, A.H. Neural induction, the default model and embryonic stem cells. Nat. Rev. Neurosci. 3, 271–280 (2002).

    Article  PubMed  CAS  Google Scholar 

  51. James, D., Levine, A.J., Besser, D. & Hemmati-Brivanlou, A. TGF-β/activin/nodal signaling is necessary for the maintenance of pluripotency in human embryonic stem cells. Development 132, 1273–1282 (2005).

    Article  CAS  PubMed  Google Scholar 

  52. Camus, A., Perea-Gomez, A., Moreau, A. & Collignon, J. Absence of Nodal signaling promotes precocious neural differentiation in the mouse embryo. Dev. Biol. 295, 743–755 (2006).

    Article  CAS  PubMed  Google Scholar 

  53. Chambers, S.M. et al. Highly efficient neural conversion of human ES and iPS cells by dual inhibition of SMAD signaling. Nat. Biotechnol. 27, 275–280 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Muguruma, K. et al. Ontogeny-recapitulating generation and tissue integration of ES cell-derived Purkinje cells. Nat. Neurosci. 13, 1171–1180 (2010).

    Article  CAS  PubMed  Google Scholar 

  55. Bouchard, M., de Caprona, D., Busslinger, M., Xu, P. & Fritzsch, B. Pax2 and Pax8 cooperate in mouse inner ear morphogenesis and innervation. BMC Dev. Biol. 10, 89 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  56. Christophorou, N.A.D., Mende, M., Lleras-Forero, L., Grocott, T. & Streit, A. Pax2 coordinates epithelial morphogenesis and cell fate in the inner ear. Dev. Biol. 345, 180–190 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Warchol, M.E. & Richardson, G.P. Expression of the Pax2 transcription factor is associated with vestibular phenotype in the avian inner ear. Dev. Neurobiol. 69, 191–202 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  58. Lysakowski, A. et al. Molecular microdomains in a sensory terminal, the vestibular calyx ending. J. Neurosci. 31, 10101–10114 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Li, A., Xue, J. & Peterson, E.H. Architecture of the mouse utricle: macular organization and hair bundle heights. J. Neurophysiol. 99, 718–733 (2008).

    Article  CAS  PubMed  Google Scholar 

  60. Li, H., Roblin, G., Liu, H. & Heller, S. Generation of hair cells by stepwise differentiation of embryonic stem cells. Proc. Natl. Acad. Sci. USA 100, 13495–13500 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Oshima, K. et al. Mechanosensitive hair cell-like cells from embryonic and induced pluripotent stem cells. Cell 141, 704–716 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Ouji, Y., Ishizaka, S., Nakamura-Uchiyama, F. & Yoshikawa, M. In vitro differentiation of mouse embryonic stem cells into inner ear hair cell-like cells using stromal cell conditioned medium. Cell Death Dis. 3, e314 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Chen, W. et al. Restoration of auditory evoked responses by human ES-cell-derived otic progenitors. Nature 490, 278–282 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Ouji, Y., Ishizaka, S., Nakamura-Uchiyama, F., Wanaka, A. & Yoshikawa, M. Induction of inner ear hair cell-like cells from Math1-transfected mouse ES cells. Cell Death Dis. 4, e700 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Brigande, J.V. & Heller, S. Quo vadis, hair cell regeneration? Nat. Neurosci. 12, 679–685 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Ding, Q. et al. A TALEN genome-editing system for generating human stem cell-based disease models. Cell Stem Cell 12, 238–251 (2013).

    Article  CAS  PubMed  Google Scholar 

  67. Ding, Q. et al. Enhanced efficiency of human pluripotent stem cell genome editing through replacing TALENs with CRISPRs. Cell Stem Cell 12, 393–394 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Trowe, M.-O. et al. Loss of Sox9 in the periotic mesenchyme affects mesenchymal expansion and differentiation, and epithelial morphogenesis during cochlea development in the mouse. Dev. Biol. 342, 51–62 (2010).

    Article  CAS  PubMed  Google Scholar 

  69. Ying, Q.-L. et al. The ground state of embryonic stem cell self-renewal. Nature 453, 519–523 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Gaboyard, S. et al. Three-dimensional culture of newborn rat utricle using an extracellular matrix promotes formation of a cyst. Neuroscience 133, 253–265 (2005).

    Article  CAS  PubMed  Google Scholar 

  71. Lerner, S.A., Matz, G.J. & Hawkins, J.E. Aminoglycoside Ototoxicity (Little Brown and Company, 1981).

  72. Yokomizo, T. et al. Whole-mount three-dimensional imaging of internally localized immunostained cells within mouse embryos. Nat. Protoc. 7, 421–431 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank A. Mikosz, D. Patel and S. Majumdar for their technical assistance and comments on the manuscript. This work was supported by US National Institutes of Health (NIH) grants RC1DC010706, R21DC012617, and R01DC013294 and an Indiana University School of Medicine Biomedical Research Grant (to E.H.). K.R.K. was supported by a Paul and Carole Stark Neurosciences Fellowship and an Indiana Clinical and Translational Science Institute Predoctoral Fellowship (NIH TL1RR025759).

Author information

Authors and Affiliations

Authors

Contributions

K.R.K. designed the protocol, made figures and wrote the manuscript. E.H. edited the manuscript.

Corresponding authors

Correspondence to Karl R Koehler or Eri Hashino.

Ethics declarations

Competing interests

K.R.K. and E.H. have disclosed these findings to the Indiana University Research and Technology Corporation, which has filed a patent to ensure broad use of the methods for studying inner-ear development and disease mechanisms. All protocols and methods remain freely available for academic and nonprofit research in perpetuity.

Integrated supplementary information

Supplementary Figure 1 Aggregate imaging chamber.

a, Typical imaging chamber constructed from two cover glasses and a silicone isolator. One aggregate (sample) is loaded in the chamber. Note the air bubble that forms during the loading process. b, Projection drawing of an aggregate imaging chamber. Note that this chamber design is an example and can be easily customized using different sized cover glasses or silicone isolators.

Supplementary information

Supplementary Figure 1

Aggregate imaging chamber. (PDF 1922 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Koehler, K., Hashino, E. 3D mouse embryonic stem cell culture for generating inner ear organoids. Nat Protoc 9, 1229–1244 (2014). https://doi.org/10.1038/nprot.2014.100

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nprot.2014.100

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing