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.

  • Article
  • Published:

Production of human spinal-cord organoids recapitulating neural-tube morphogenesis

Abstract

Human spinal-cord-like tissues induced from human pluripotent stem cells are typically insufficiently mature and do not mimic the morphological features of neurulation. Here, we report a three-dimensional culture system and protocol for the production of human spinal-cord-like organoids (hSCOs) recapitulating the neurulation-like tube-forming morphogenesis of the early spinal cord. The hSCOs exhibited neurulation-like tube-forming morphogenesis, cellular differentiation into the major types of spinal-cord neurons as well as glial cells, and mature synaptic functional activities, among other features of the development of the spinal cord. We used the hSCOs to screen for antiepileptic drugs that can cause neural-tube defects. hSCOs may also facilitate the study of the development of the human spinal cord and the modelling of diseases associated with neural-tube defects.

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

Fig. 1: Generation of hSCOs recapitulating neural tube morphogenesis.
Fig. 2: Transcriptome profiling of hSCOs.
Fig. 3: Acquisition of spinal-cord-like cell fate after long-term culture of hSCOs.
Fig. 4: Electrophysiological analysis and pharmacological response in hSCOs.
Fig. 5: Size and bFGF duration-dependent tube morphogenesis in hSCOs.
Fig. 6: Modelling of antiepileptic drug-induced neural tube defects in hSCOs using deep learning-based classification.

Similar content being viewed by others

Data availability

The main data supporting the results in this study are available within the paper and its Supplementary Information. Gene expression data are available in the Gene Expression Omnibus (GEO) under accession numbers GSE196573 (for single-cell RNA-sequencing data) and GSE157696 (for microarray data). Source data are provided with this paper.

Code availability

The code for training the deep learning models in this study is available at https://github.com/im-namwon/stemcell-classification.

References

  1. Smith, J. L. & Schoenwolf, G. C. Neurulation: coming to closure. Trends Neurosci. 20, 510–517 (1997).

    Article  CAS  PubMed  Google Scholar 

  2. Jankowska, E. Spinal interneuronal systems: identification, multifunctional character and reconfigurations in mammals. J. Physiol. 533, 31–40 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Lu, D. C., Niu, T. & Alaynick, W. A. Molecular and cellular development of spinal cord locomotor circuitry. Front. Mol. Neurosci. 8, 25 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  4. Greene, N. D., Stanier, P. & Copp, A. J. Genetics of human neural tube defects. Hum. Mol. Genet. 18, R113–R129 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Loeken, M. R. Current perspectives on the causes of neural tube defects resulting from diabetic pregnancy. Am. J. Med. Genet. C 135, 77–87 (2005).

  6. Matok, I. et al. Exposure to folic acid antagonists during the first trimester of pregnancy and the risk of major malformations. Br. J. Clin. Pharmacol. 68, 956–962 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Mølgaard-Nielsen, D. & Hviid, A. Newer-generation antiepileptic drugs and the risk of major birth defects. JAMA 305, 1996–2002 (2011).

    Article  PubMed  Google Scholar 

  8. Warmflash, A., Sorre, B., Etoc, F., Siggia, E. D. & Brivanlou, A. H. A method to recapitulate early embryonic spatial patterning in human embryonic stem cells. Nat. Methods 11, 847–854 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Haremaki, T. et al. Self-organizing neuruloids model developmental aspects of Huntington’s disease in the ectodermal compartment. Nat. Biotechnol. 37, 1198–1208 (2019).

    Article  CAS  PubMed  Google Scholar 

  10. Moris, N. et al. An in vitro model of early anteroposterior organization during human development. Nature 582, 410–415 (2020).

  11. Kim, J., Koo, B.-K. & Knoblich, J. A. Human organoids: model systems for human biology and medicine. Nat. Rev. Mol. Cell Biol. 21, 571–584 (2020).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  13. Qian, X. et al. Brain-region-specific organoids using mini-bioreactors for modeling ZIKV exposure. Cell 165, 1238–1254 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Quadrato, G. et al. Cell diversity and network dynamics in photosensitive human brain organoids. Nature 545, 48 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Paşca, A. M. et al. Functional cortical neurons and astrocytes from human pluripotent stem cells in 3D culture. Nat. Methods 12, 671 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Kadoshima, T. et al. Self-organization of axial polarity, inside-out layer pattern, and species-specific progenitor dynamics in human ES cell–derived neocortex. Proc. Natl Acad. Sci. USA 110, 20284–20289 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Meinhardt, A. et al. 3D reconstitution of the patterned neural tube from embryonic stem cells. Stem Cell Rep. 3, 987–999 (2014).

    Article  Google Scholar 

  18. Ogura, T., Sakaguchi, H., Miyamoto, S. & Takahashi, J. Three-dimensional induction of dorsal, intermediate and ventral spinal cord tissues from human pluripotent stem cells. Development 145, dev162214 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  19. Hor, J. H. et al. Cell cycle inhibitors protect motor neurons in an organoid model of spinal muscular atrophy. Cell Death Dis. 9, 1–12 (2018).

    Article  CAS  Google Scholar 

  20. Martins, J.-M. F. et al. Self-organizing 3D human trunk neuromuscular organoids. Cell Stem Cell 26, 172–186.e176 (2020).

    Article  CAS  Google Scholar 

  21. Rifes, P. et al. Modeling neural tube development by differentiation of human embryonic stem cells in a microfluidic WNT gradient. Nat. Biotechnol. 38, 1265–1273 (2020).

  22. Quadrato, G., Brown, J. & Arlotta, P. The promises and challenges of human brain organoids as models of neuropsychiatric disease. Nat. Med. 22, 1220 (2016).

    Article  CAS  PubMed  Google Scholar 

  23. Velasco, S. et al. Individual brain organoids reproducibly form cell diversity of the human cerebral cortex. Nature 570, 523–527 (2019).

  24. Denham, M. et al. Multipotent caudal neural progenitors derived from human pluripotent stem cells that give rise to lineages of the central and peripheral nervous system. Stem Cells 33, 1759–1770 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Anderson, M. J. et al. TCreERT2, a transgenic mouse line for temporal control of Cre-mediated recombination in lineages emerging from the primitive streak or tail bud. PLoS ONE 8, e62479 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Nagai, T. et al. The expression of the mouse Zic1, Zic2, and Zic3 gene suggests an essential role for Zic genes in body pattern formation. Dev. Biol. 182, 299–313 (1997).

    Article  CAS  PubMed  Google Scholar 

  27. Pyrgaki, C., Trainor, P., Hadjantonakis, A. K. & Niswander, L. Dynamic imaging of mammalian neural tube closure. Dev. Biol. 344, 941–947 (2010).

    Article  CAS  PubMed  Google Scholar 

  28. Aaku-Saraste, E., Hellwig, A. & Huttner, W. B. Loss of occludin and functional tight junctions, but not ZO-1, during neural tube closure—remodeling of the neuroepithelium prior to neurogenesis. Dev. Biol. 180, 664–679 (1996).

    Article  CAS  PubMed  Google Scholar 

  29. Nishimura, T., Honda, H. & Takeichi, M. Planar cell polarity links axes of spatial dynamics in neural-tube closure. Cell 149, 1084–1097 (2012).

    Article  CAS  PubMed  Google Scholar 

  30. Stuart, T. et al. Comprehensive integration of single-cell data. Cell 177, 1888–1902.e1821 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Delile, J. et al. Single cell transcriptomics reveals spatial and temporal dynamics of gene expression in the developing mouse spinal cord. Development 146, dev173807 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Floyd, T. L., Dai, Y. & Ladle, D. R. Characterization of calbindin D28k expressing interneurons in the ventral horn of the mouse spinal cord. Dev. Dyn. 247, 185–193 (2018).

    Article  CAS  PubMed  Google Scholar 

  33. Dale, N. Reciprocal inhibitory interneurones in the Xenopus embryo spinal cord. J. Physiol. 363, 61–70 (1985).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Shin, H. et al. Multifunctional multi-shank neural probe for investigating and modulating long-range neural circuits in vivo. Nat. Commun. 10, 3777 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. Hanson, M. G. & Landmesser, L. T. Characterization of the circuits that generate spontaneous episodes of activity in the early embryonic mouse spinal cord. J. Neurosci. 23, 587–600 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Zafeiriou, M.-P. et al. Developmental GABA polarity switch and neuronal plasticity in bioengineered neuronal organoids. Nat. Commun. 11, 3791 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Greene, N. D. & Copp, A. J. Neural tube defects. Annu. Rev. Neurosci. 37, 221–242 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Agopian, A., Tinker, S. C., Lupo, P. J., Canfield, M. A. & Mitchell, L. E. Proportion of neural tube defects attributable to known risk factors. Birth Defects Res. A 97, 42–46 (2013).

    Article  CAS  Google Scholar 

  39. Weston, J. et al. Monotherapy treatment of epilepsy in pregnancy: congenital malformation outcomes in the child. Cochrane Database Syst. Rev. 11, CD010224 (2016).

  40. Hughes, A., Greene, N. D., Copp, A. J. & Galea, G. L. Valproic acid disrupts the biomechanics of late spinal neural tube closure in mouse embryos. Mech. Dev. 149, 20–26 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Sahni, G. et al. A micropatterned human-specific neuroepithelial tissue for modeling gene and drug‐induced neurodevelopmental defects. Adv. Sci. 8, 2001100 (2021).

    Article  CAS  Google Scholar 

  42. Kawada, J. et al. Generation of a motor nerve organoid with human stem cell-derived neurons. Stem Cell Rep. 9, 1441–1449 (2017).

    Article  Google Scholar 

  43. Sternfeld, M. J. et al. Speed and segmentation control mechanisms characterized in rhythmically-active circuits created from spinal neurons produced from genetically-tagged embryonic stem cells. eLife 6, e21540 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  44. Zheng, Y. et al. Dorsal-ventral patterned neural cyst from human pluripotent stem cells in a neurogenic niche. Sci. Adv. 5, eaax5933 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Davidson, B. P., Kinder, S. J., Steiner, K., Schoenwolf, G. C. & Tam, P. P. Impact of node ablation on the morphogenesis of the body axis and the lateral asymmetry of the mouse embryo during early organogenesis. Dev. Biol. 211, 11–26 (1999).

    Article  CAS  PubMed  Google Scholar 

  46. Stemple, D. L. Structure and function of the notochord: an essential organ for chordate development. Development 132, 2503–2512 (2005).

    Article  CAS  PubMed  Google Scholar 

  47. Moury, J. D. & Schoenwolf, G. C. Cooperative model of epithelial shaping and bending during avian neurulation: autonomous movements of the neural plate, autonomous movements of the epidermis, and interactions in the neural plate/epidermis transition zone. Dev. Dyn. 204, 323–337 (1995).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  49. Lowery, L. A. & Sive, H. Strategies of vertebrate neurulation and a re-evaluation of teleost neural tube formation. Mech. Dev. 121, 1189–1197 (2004).

    Article  CAS  PubMed  Google Scholar 

  50. Yoon, S.-J. et al. Reliability of human cortical organoid generation. Nat. Met. 16, 75–78 (2019).

    Article  CAS  Google Scholar 

  51. Bakkum, D. J. et al. Parameters for burst detection. Front. Comput. Neurosci. 7, 193 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  52. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    Article  CAS  PubMed  Google Scholar 

  53. Butler, A., Hoffman, P., Smibert, P., Papalexi, E. & Satija, R. Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat. Biotechnol. 36, 411–420 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Hafemeister, C. & Satija, R. Normalization and variance stabilization of single-cell RNA-seq data using regularized negative binomial regression. Genome Biol. 20, 1–15 (2019).

    Article  CAS  Google Scholar 

  55. Sakar, M. S. et al. Formation and optogenetic control of engineered 3D skeletal muscle bioactuators. Lab Chip 12, 4976–4985 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We would like to thank the Korea Basic Science Institute, Korea Brain Research Institute, K.-S. Yang and J. Na for their technical support. We especially thank SCREEN Holdings (Kyoto, Japan) and Kim & Friends (Seoul, Republic of Korea) for technical support utilizing the OCT instrument, Cell3iMager Estier. We also thank J. Hosung (Yonsei University) for his critical comments. This research was supported by the Brain Research Program through the National Research Foundation (NRF), which is funded by the Korean Ministry of Science, ICT and Future Planning (NRF-2015M3C7A1028790, NRF-2017M3C7A1047654, NRF-2017M3A9B3061308 and NRF-2021M3E5D9021368).

Author information

Authors and Affiliations

Authors

Contributions

J.-H.L. and W.S. conceived the project and designed the experiments. J.-H.L. performed most of the experiments, and analysed the data. H.S. performed electrophysiological experiments and analysed the data. M.R.S., H.J.K., S.-H.P., J.H.K., M.K. and T.H.K. performed the experiments. N.L. performed machine learning experiments. S.C. performed and interpreted bioinformatics analysis. J.W.K., M.-R.S., S.-H.K., D.W.H., S.L., S.-Y.C., I.J.R., H.K., D.G. and I.-J.C. provided the resource. S.L., S.-Y.C., D.G., I.-J.C. and W.S. designed the study. W.S. and J.H.L. wrote the manuscript. W.S. supervised and administered this project. All authors discussed the results and contributed to the revision of the manuscript.

Corresponding author

Correspondence to Woong Sun.

Ethics declarations

Competing interests

N.L. is employed by InterMinds. The authors declare no other competing interests.

Peer review

Peer review information

Nature Biomedical Engineering thanks Agnete Kirkeby, Matthias Lutolf and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Generation of caudal NSCs from hiPSCs in 2D.

a. Caudal induction (SB + CHIR) of hiPSCs on 2D culture showed the induction of the NMP markers SOX2/BraT and CDX2, and then subsequent leading to the conversion of hiPSCs into N-cad positive neural stem cells. b. High-magnification images of NMPs on induction day 2. These cells showed co-expression of the representative NMP markers (SOX2, BraT and CDX2). c. Real-time PCR profiles of gene expression for neuromesoderm (BraT, NKX1.2, CDX2 and SOX2), pluripotency marker (OCT4 and NANOG), mesendoderm (MIXL1), mesoderm (TBX6), neuroectoderm (SOX1 and PAX6), and rostral neuroectoderm (OTX2). d. The E- to N-cadherin switch was present during 2D induction. Immunofluorescence analysis of E-cadherin (red), N-cadherin (green), and NANOG (white). Nuclei were counterstained with Hoechst (blue). e. Relative expression of the E- and N- cadherin mRNAs, depending on the days of differentiation. f. The maintenance of apical polarity from 2D to 3D culture. During the 3D conversion process, surface neuroepithelia cells re-established their polarity within 24 hours. Apical polarity was visualised with ZO1 immunolabeling (red). Nuclei were counterstained with Hoechst (blue). g. The pseudostratified morphology of NE cells on day 4 (bFGF, day 1). Note that the internal neural stem cells (SOX2 + ) exhibit distinct cell morphology. Cell morphologies were visualised with a small fraction of GFP-labelled cells initially mixed with non-labelled hPSCs. h. Absence of mesodermal cells in 3D spheres. The neuroepithelia cells were visualised with SOX2 (green) and the mesodermal cells were visualised with BraT or TBX6 (red) staining. Nuclei were counterstained with Hoechst (blue). Although BraT expression was observed in small number of organoids at early stages of culture (day 4), they were eventually disappeared (day 7). In addition, we failed to detect any TBX6-expressing mesodermal cells. These data suggested that our induction condition exclusively drove cells toward neural fates. Data were collected and analysed from three independent experiment samples. Data are expressed as mean ± SEM. Standard one-way ANOVA with Tukey’s multiple comparison test was used for multiple comparisons (c and e). All scale bars, 50 μm.

Source data

Extended Data Fig. 2 hSCOs mimicking neurulation morphogenesis in vivo.

a. Morphology of neural folds at different stages in epidermal fibroblast-derived hiPSCs (left) and H9 hESCs (right). b. Immunofluorescence staining showed the absence of floor plate cells (FOXA2), roof plate cells (LMX1A), and neural crest cells (SLUG, p75, SOX10 and FOXD3). c. Absence of non-neural tissue components in the hSCOs. The non-neural epithelium was visualised with E-cadherin (green) and the mesodermal tissue was visualised with BraT (red) staining. Developing mouse neural tube (bottom) is shown as a control. d. High-magnification image of the planar cell polarity (PCP) region. The right-bottom image shows traces of pMLC and ZO1 expression. e. Quantification of PCP with angular analysis of pMLC linearity (left) and ZO1 linearity (right). To measure the angles of pMLC and ZO1 cables, we selected a line linking two or more cells. Compared to ZO1 cables, the pMLC cables tended to align laterally. Data are represented as mean ± SD. f. The ratio of the medio-lateral (M-L) like axis (from 45 to 135 degrees) to the anterior-posterior (A-P) like axis (from 0 to 45 and from 135 to 180 degrees). Data are represented as mean ± SD. Unpaired student’s t-test was used for comparing two groups. g. Scanning electron microscopy (SEM) images of hSCOs at different stages of development. Note that the surfaces of the first spheroids were relatively smooth with strong tight junctions, while neural tube stage hSCOs had rough surfaces. All scale bars, 50 μm.

Source data

Extended Data Fig. 3 Characterization of the neural tube in hSCOs.

a. Interkinetic nuclear migration (IKNM) in hSCO on day 15. In neural tube, S-phase cells probed by EdU (green) incorporation were localised at the basal side, while M-phase cells labelled by phospho-histone H3 (p-H3, red) were located on the apical side of tube. The neural tube structure was visualised with SOX2 (blue). Scale bar, 100 µm. b. Representative images of IKNM in the neural tube. EdU pulse (15-min)-labelled cells moved toward the apical side over time, and then back to basal side. c. Quantification of p-H3+EdU+ cells per total p-H3+ cells in the neural tube. Data are expressed as mean ± SD. The number of analysed organoids (n) were summarised in Supplementary Table 5. d. Mitotic radial glia labelled with phospho-vimentin (red) were located on the apical side of the neural tube. The nuclei were counterstained with Hoechst (blue). e. The presence of primary cilia at apical side of the neural tube-like structure. Immunofluorescence for ARL13B (red) and γ-tubulin (green) detected the primary cilia and basal bodies, respectively. The nuclei were counterstained with Hoechst (blue). f. Double immunostaining for the neural stem cell marker SOX2 (red) and the neuroblast marker DCX (green). The outside of the neural tube was surrounded by early differentiated neuronal cells. All scale bars, 20 μm; except panels (a).

Source data

Extended Data Fig. 4 Effects of perturbation of apico-basal polarity on neural tube morphogenesis.

a. Treatment scheme of Y-27632 (Rock inhibitor; 10 μM) for panels (b). b. The effects of Y-27632 on the establishment of the neural plate. Note that the Y-27632 treatments disrupted apical localisation of ZO1. c. Treatment scheme of Y-27632 for panels (d). d. The effects of Y-27632 at the neural folding-stage. Note that the neural folding was not induced by the Y-27632 treatments, and the organoids exhibited round morphology. e. Treatment scheme for Matrigel-embedding experiments on panels (f). f. Matrigel embedding at the early neural plate-stage rapidly reversed the apical polarity, and promoted cavitation (by 72 hr). g. Treatment scheme for Matrigel-embedding experiments on panels (h-j). h. Matrigel embedding at the neural folding-stage also resulted in ventricle-like morphogenesis, as previously reported with forebrain organoids. i. 3D structure of the cavities in the Matrigel embedded-hSCO. The cavities were labelled with ZO1 staining. Note that the control hSCO exhibited highly connected central canal-like structure, and Matrigel embedded-hSCO exhibited short and isolated follicle-like structure12,13. j. Quantification of total cavities in individual organoids. Data are expressed as mean ± SD. Unpaired student’s t-test was used for comparing two groups. The number of analysed organoids (n) were summarised in Supplementary Table 5. The apical polarity at the neural plate or neural folding-stage was visualised with ZO1 (red) and the nuclei were counterstained with Hoechst (blue) in all panels. All scale bars, 50 μm.

Source data

Extended Data Fig. 5 Changes in the patterns of neural activities in hSCOs.

a-d. Representative heatmaps showing neural activities at Day 24 (a), Day 46 (b), Day 72 (c), and Day 140 (d). e-h. Representative cross-correlation matrices showing synchronisation between signal-recorded electrodes at Day 24 (e), Day 46 (f), Day 72 (g), and Day 140 (h). i-l. Changes in the patterns of burst activity as the hSCOs mature. Bar graphs represent mean ± SD in total spike number in burst activities (i), burst duration (j), inter burst interval (IBI) (k), and inter spike interval (ISI) in burst activities (l). Unpaired student’s t-test was used for comparing two groups. The number of analysed organoids (n) and the number of experiments (N) were summarised in Supplementary Table 5. m. Representative transient plot showing electrically evoked activities in matured hSCOs and expanded plots of the first and fifth stimulus.

Source data

Supplementary information

Supplementary Information

Supplementary figs., tables and video captions.

Reporting Summary

Peer Review Information

Supplementary Table 5

Number of analysed organoids (n) and number of experiments (N).

Supplementary Datasets

Source data for Supplementary Figs. 11, 12 and 14.

Supplementary Video 1

Live imaging of 3D conversion. Video related to Fig. 1a.

Supplementary Video 2

Live imaging of the hSCOs exhibiting neural tube closure. Video related to Fig. 1b.

Supplementary Video 3

Time-course images of neurulation-like morphogenesis in hSCOs. Video related to Fig. 1c.

Supplementary Video 4

Morphology of hinge cells. Video related to Fig. 1g.

Supplementary Video 5

Zippering-like morphogenesis in hSCO. Video related to Fig. 1l.

Supplementary Video 6

3D architecture of neural tube-stage hSCO. Video related to Fig. 1m.

Supplementary Video 7

Core-to-shell organization of hSCOs. Video related to Fig. 3a.

Supplementary Video 8

Comparison between dispase- and accutase-based protocols for 3D conversion. Video related to Supplementary Fig. 11.

Supplementary Video 9

3D neural tube morphology in 6 AEDs-treated hSCOs. Video related to Fig. 6e.

Source data

Source Data Fig. 3

Source data for Fig. 3.

Source Data Fig. 4

Source data for Fig. 4.

Source Data Fig. 5

Source data for Fig. 5.

Source Data Fig. 6

Source data and statistics for Fig. 6.

Source Data Extended Data Fig. 1

Source data for Extended Data Fig. 1.

Source Data Extended Data Fig. 2

Source data for Extended Data Fig. 2.

Source Data Extended Data Fig. 3

Source data for Extended Data Fig. 3.

Source Data Extended Data Fig. 4

Source data for Extended Data Fig. 4.

Source Data Extended Data Fig. 5

Source data for Extended Data Fig. 5.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lee, JH., Shin, H., Shaker, M.R. et al. Production of human spinal-cord organoids recapitulating neural-tube morphogenesis. Nat. Biomed. Eng 6, 435–448 (2022). https://doi.org/10.1038/s41551-022-00868-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41551-022-00868-4

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research