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.

  • Technical Report
  • Published:

Differentiation and maturation of oligodendrocytes in human three-dimensional neural cultures

Abstract

Investigating human oligodendrogenesis and the interaction of oligodendrocytes with neurons and astrocytes would accelerate our understanding of the mechanisms underlying white matter disorders. However, this is challenging because of the limited accessibility of functional human brain tissue. Here, we developed a new differentiation method of human induced pluripotent stem cells to generate three-dimensional brain organoids that contain oligodendrocytes as well as neurons and astrocytes, called human oligodendrocyte spheroids. We found that oligodendrocyte lineage cells derived in human oligodendrocyte spheroids transitioned through developmental stages similar to primary human oligodendrocytes and that the migration of oligodendrocyte lineage cells and their susceptibility to lysolecithin exposure could be captured by live imaging. Moreover, their morphology changed as they matured over time in vitro and started myelinating neurons. We anticipate that this method can be used to study oligodendrocyte development, myelination, and interactions with other major cell types in the CNS.

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: Characterization of hOLS derived from hiPS cell lines.
Fig. 2: Transcriptional comparison of hOLS oligodendrocyte lineage cells to primary tissue cells.
Fig. 3: Developmental trajectory and expression of disease-related genes in hOLS.
Fig. 4: Oligodendrocyte maturation in hOLS.
Fig. 5: Oligodendrocyte–neuron interaction and myelination in hOLS.

Similar content being viewed by others

Data availability

Gene expression data have been deposited in the Gene Expression Omnibus under accession number GSE115011. The data that support the findings of this study are available on reasonable request from the corresponding author.

References

  1. Nave, K. A. Myelination and the trophic support of long axons. Nat. Rev. Neurosci. 11, 275–283 (2010).

    Article  CAS  Google Scholar 

  2. Menichella, D. M. et al. Genetic and physiological evidence that oligodendrocyte gap junctions contribute to spatial buffering of potassium released during neuronal activity. J. Neurosci. 26, 10984–10991 (2006).

    Article  CAS  Google Scholar 

  3. Fünfschilling, U. et al. Glycolytic oligodendrocytes maintain myelin and long-term axonal integrity. Nature 485, 517–521 (2012).

    Article  Google Scholar 

  4. Dai, X. et al. The trophic role of oligodendrocytes in the basal forebrain. J. Neurosci. 23, 5846–5853 (2003).

    Article  CAS  Google Scholar 

  5. Orthmann-Murphy, J. L., Freidin, M., Fischer, E., Scherer, S. S. & Abrams, C. K. Two distinct heterotypic channels mediate gap junction coupling between astrocyte and oligodendrocyte connexins. J. Neurosci. 27, 13949–13957 (2007).

    Article  CAS  Google Scholar 

  6. Moore, C. S., Abdullah, S. L., Brown, A., Arulpragasam, A. & Crocker, S. J. How factors secreted from astrocytes impact myelin repair. J. Neurosci. Res. 89, 13–21 (2011).

    Article  CAS  Google Scholar 

  7. Simons, M. & Trajkovic, K. Neuron-glia communication in the control of oligodendrocyte function and myelin biogenesis. J. Cell Sci. 119, 4381–4389 (2006).

    Article  CAS  Google Scholar 

  8. Barres, B. A., Schmid, R., Sendnter, M. & Raff, M. C. Multiple extracellular signals are required for long-term oligodendrocyte survival. Development 118, 283–295 (1993).

    CAS  PubMed  Google Scholar 

  9. Lin, S. C. & Bergles, D. E. Synaptic signaling between GABAergic interneurons and oligodendrocyte precursor cells in the hippocampus. Nat. Neurosci. 7, 24–32 (2004).

    Article  CAS  Google Scholar 

  10. Bergles, D. E., Roberts, J. D., Somogyi, P. & Jahr, C. E. Glutamatergic synapses on oligodendrocyte precursor cells in the hippocampus. Nature 405, 187–191 (2000).

    Article  CAS  Google Scholar 

  11. Hardy, R. & Reynolds, R. Neuron-oligodendroglial interactions during central nervous system development. J. Neurosci. Res. 36, 121–126 (1993).

    Article  CAS  Google Scholar 

  12. van der Knaap, M. S., Pronk, J. C. & Scheper, G. C. Vanishing white matter disease. Lancet Neurol. 5, 413–423 (2006).

    Article  Google Scholar 

  13. Wolswijk, G. Oligodendrocyte survival, loss and birth in lesions of chronic-stage multiple sclerosis. Brain 123, 105–115 (2000).

    Article  Google Scholar 

  14. Douvaras, P. & Fossati, V. Generation and isolation of oligodendrocyte progenitor cells from human pluripotent stem cells. Nat. Protoc. 10, 1143–1154 (2015).

    Article  CAS  Google Scholar 

  15. Numasawa-Kuroiwa, Y. et al. Involvement of ER stress in dysmyelination of Pelizaeus–Merzbacher disease with PLP1 missense mutations shown by iPSC-derived oligodendrocytes. Stem Cell Rep. 2, 648–661 (2014).

    Article  CAS  Google Scholar 

  16. Stacpoole, S. R. et al. High yields of oligodendrocyte lineage cells from human embryonic stem cells at physiological oxygen tensions for evaluation of translational biology. Stem Cell Rep. 1, 437–450 (2013).

    Article  CAS  Google Scholar 

  17. Wang, S. et al. Human iPSC-derived oligodendrocyte progenitor cells can myelinate and rescue a mouse model of congenital hypomyelination. Cell Stem Cell 12, 252–264 (2013).

    Article  CAS  Google Scholar 

  18. Hogberg, H. T. et al. Toward a 3D model of human brain development for studying gene/environment interactions. Stem Cell Res. Ther. 4(Suppl 1), S4 (2013).

    Article  Google Scholar 

  19. Pașca, S. P. The rise of three-dimensional human brain cultures. Nature 553, 437–445 (2018).

    PubMed  Google Scholar 

  20. Paşca, A. M. et al. Functional cortical neurons and astrocytes from human pluripotent stem cells in 3D culture. Nat. Methods 12, 671–678 (2015).

    Article  Google Scholar 

  21. Birey, F. et al. Assembly of functionally integrated human forebrain spheroids. Nature 545, 54–59 (2017).

    Article  CAS  Google Scholar 

  22. Sloan, S. A., Andersen, J., Pașca, A. M., Birey, F. & Pașca, S. P. Generation and assembly of human brain region-specific three-dimensional cultures. Nat. Protoc. 13, 2062–2085 (2018).

    Article  CAS  Google Scholar 

  23. Darmanis, S. et al. A survey of human brain transcriptome diversity at the single cell level. Proc. Natl Acad. Sci. USA 112, 7285–7290 (2015).

    Article  CAS  Google Scholar 

  24. Sloan, S. A. et al. Human astrocyte maturation captured in 3D cerebral cortical spheroids derived from pluripotent stem cells. Neuron 95, 779–790.e6 (2017).

    Article  CAS  Google Scholar 

  25. van der Maaten, L. & Hinton, G. Visualizing data using t-SNE. J. Mach. Learn. Res. 9, 2579–2605 (2008).

    Google Scholar 

  26. Trapnell, C. et al. The dynamics and regulators of cell fate decisions are revealed by pseudotemporal ordering of single cells. Nat. Biotechnol. 32, 381–386 (2014).

    Article  CAS  Google Scholar 

  27. Zhang, Y. et al. Purification and characterization of progenitor and mature human astrocytes reveals transcriptional and functional differences with mouse. Neuron 89, 37–53 (2016).

    Article  CAS  Google Scholar 

  28. Crow, Y. J. et al. Mutations in genes encoding ribonuclease H2 subunits cause Aicardi–Goutières syndrome and mimic congenital viral brain infection. Nat. Genet. 38, 910–916 (2006).

    Article  CAS  Google Scholar 

  29. Gomez-Ospina, N. Arylsulfatase A deficiency. in GeneReviews (eds. Adam, M. P. et al.) (Univ. Washington, Seattle, 2006).

  30. Austin, J. et al. Studies in globoid (Krabbe) leukodystrophy (GLD). V. Controlled enzymic studies in ten human cases. Arch. Neurol. 23, 502–512 (1970).

    Article  CAS  Google Scholar 

  31. Bergles, D. E. & Richardson, W. D. Oligodendrocyte development and plasticity. Cold Spring Harb. Perspect. Biol. 8, a020453 (2015).

    Article  Google Scholar 

  32. Pol, S. U. et al. Sox10-MCS5 enhancer dynamically tracks human oligodendrocyte progenitor fate. Exp. Neurol. 247, 694–702 (2013).

    Article  CAS  Google Scholar 

  33. Sontheimer, H., Trotter, J., Schachner, M. & Kettenmann, H. Channel expression correlates with differentiation stage during the development of oligodendrocytes from their precursor cells in culture. Neuron 2, 1135–1145 (1989).

    Article  CAS  Google Scholar 

  34. Livesey, M. R. et al. Maturation and electrophysiological properties of human pluripotent stem cell-derived oligodendrocytes. Stem Cells 34, 1040–1053 (2016).

    Article  CAS  Google Scholar 

  35. Káradóttir, R. & Attwell, D. Neurotransmitter receptors in the life and death of oligodendrocytes. Neuroscience 145, 1426–1438 (2007).

    Article  Google Scholar 

  36. Káradóttir, R., Cavelier, P., Bergersen, L. H. & Attwell, D. NMDA receptors are expressed in oligodendrocytes and activated in ischaemia. Nature 438, 1162–1166 (2005).

    Article  Google Scholar 

  37. Birgbauer, E., Rao, T. S. & Webb, M. Lysolecithin induces demyelination in vitro in a cerebellar slice culture system. J. Neurosci. Res. 78, 157–166 (2004).

    Article  CAS  Google Scholar 

  38. Blakemore, W. F., Eames, R. A., Smith, K. J. & McDonald, W. I. Remyelination in the spinal cord of the cat following intraspinal injections of lysolecithin. J. Neurol. Sci. 33, 31–43 (1977).

    Article  CAS  Google Scholar 

  39. Hall, S. M. The effect of injections of lysophosphatidyl choline into white matter of the adult mouse spinal cord. J. Cell Sci. 10, 535–546 (1972).

    CAS  PubMed  Google Scholar 

  40. Amin, N. D. & Paşca, S. P. Building models of brain disorders with three-dimensional organoids. Neuron 100, 389–405 (2018).

    Article  CAS  Google Scholar 

  41. Di Lullo, E. & Kriegstein, A. R. The use of brain organoids to investigate neural development and disease. Nat. Rev. Neurosci. 18, 573–584 (2017).

    Article  Google Scholar 

  42. Yoon, S. et al. Reliability of human cortical organoid generation. Nat. Methods 16, 75–78 (2019).

    Article  CAS  Google Scholar 

  43. Volpe, J. J. The encephalopathy of prematurity – brain injury and impaired brain development inextricably intertwined. Semin. Pediatr. Neurol. 16, 167–178 (2009).

    Article  Google Scholar 

  44. Paşca, S. P. et al. Using iPSC-derived neurons to uncover cellular phenotypes associated with Timothy syndrome. Nat. Med. 17, 1657–1662 (2011).

    Article  Google Scholar 

  45. Yazawa, M. et al. Using induced pluripotent stem cells to investigate cardiac phenotypes in Timothy syndrome. Nature 471, 230–234 (2011).

    Article  CAS  Google Scholar 

  46. Gallego Romero, I. et al. A panel of induced pluripotent stem cells from chimpanzees: a resource for comparative functional genomics. eLife 4, e07103 (2015).

    Article  Google Scholar 

  47. Marchetto, M. C. et al. Non-cell-autonomous effect of human SOD1 G37R astrocytes on motor neurons derived from human embryonic stem cells. Cell Stem Cell 3, 649–657 (2008).

    Article  CAS  Google Scholar 

  48. Xue, Y. et al. Sequential regulatory loops as key gatekeepers for neuronal reprogramming in human cells. Nat. Neurosci. 19, 807–815 (2016).

    Article  CAS  Google Scholar 

  49. The HD iPSC Consortium. Developmental alterations in Huntington’s disease neural cells and pharmacological rescue in cells and mice. Nat. Neurosci. 20, 648–660 (2017).

  50. Renner, M. et al. Self-organized developmental patterning and differentiation in cerebral organoids. EMBO J. 36, 1316–1329 (2017).

    Article  CAS  Google Scholar 

  51. Kang, S. M. et al. Efficient induction of oligodendrocytes from human embryonic stem cells. Stem Cells 25, 419–424 (2007).

    Article  CAS  Google Scholar 

  52. Deborde, S. et al. Schwann cells induce cancer cell dispersion and invasion. J. Clin. Invest. 126, 1538–1554 (2016).

    Article  Google Scholar 

  53. Xiao, D. et al. Direct reprogramming of fibroblasts into neural stem cells by single non-neural progenitor transcription factor Ptf1a. Nat. Commun. 9, 2865 (2018).

    Article  Google Scholar 

  54. Clarke, K. E. et al. A robust and reproducible human pluripotent stem cell derived model of neurite outgrowth in a three-dimensional culture system and its application to study neurite inhibition. Neurochem. Int. 106, 74–84 (2017).

    Article  CAS  Google Scholar 

  55. Greber, B. et al. FGF signalling inhibits neural induction in human embryonic stem cells. EMBO J. 30, 4874–4884 (2011).

    Article  CAS  Google Scholar 

  56. Amin, H. et al. Electrical responses and spontaneous activity of human iPS-derived neuronal networks characterized for 3-month culture with 4096-electrode arrays. Front. Neurosci. 10, 121 (2016).

    Article  CAS  Google Scholar 

  57. Picelli, S. et al. Full-length RNA-seq from single cells using Smart-seq2. Nat. Protoc. 9, 171–181 (2014).

    Article  CAS  Google Scholar 

  58. Risso, D., Ngai, J., Speed, T. P. & Dudoit, S. Normalization of RNA-seq data using factor analysis of control genes or samples. Nat. Biotechnol. 32, 896–902 (2014).

    Article  CAS  Google Scholar 

  59. Penna, I. et al. Selection of candidate housekeeping genes for normalization in human postmortem brain samples. Int. J. Mol. Sci. 12, 5461–5470 (2011).

    Article  CAS  Google Scholar 

  60. Fan, J. et al. Characterizing transcriptional heterogeneity through pathway and gene set overdispersion analysis. Nat. Methods 13, 241–244 (2016).

    Article  CAS  Google Scholar 

  61. Deverman, B. E. et al. Cre-dependent selection yields AAV variants for widespread gene transfer to the adult brain. Nat. Biotechnol. 34, 204–209 (2016).

    Article  CAS  Google Scholar 

  62. Thomas, C. A. et al. Modeling of TREX1-dependent autoimmune disease using human stem cells highlights L1 accumulation as a source of neuroinflammation. Cell Stem Cell 21, 319–331.e8 (2017).

    Article  CAS  Google Scholar 

  63. Mariani, J. et al. FOXG1-dependent dysregulation of GABA/glutamate neuron differentiation in autism spectrum disorders. Cell 162, 375–390 (2015).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This paper is dedicated to the memory of our wonderful colleague and mentor Ben A. Barres. We thank B.A. Barres, B. Zuchero, and members of the Pasca laboratory for scientific input, J. Perrino (Stanford Cell Sciences Imaging Facility) for support with electron microscopy, as well as F. Sim (University of Buffalo) for providing the Sox10-MCS5::eGFP plasmid. This work was supported by the US National Institutes of Health BRAINS Award (MH107800), the MQ Fellow Award, the NYSCF Robertson Stem Cell Investigator Award, the Stanford Wu Tsai Neurosciences Institute’s Brain Rejuvenation Project and the Human Brain Organogenesis Project, the Kwan Research Fund and the California Institute of Regenerative Medicine, the Child Health Research Institute Pilot Award, and the NARSAD Independent Investigator Award from the Brain and Behavior Research Foundation (to S.P.P.); the National Science Foundation Graduate Research Fellowship and the Bio-X Stanford Interdisciplinary Graduate Fellowship (to R.M.M.); Stanford Medicine’s Dean’s Fellowship (to Y.M.); and NIMH T32GM007365, F30MH106261, and Bio-X Predoctoral Fellowship (to or supporting S.A.S.).

Author information

Authors and Affiliations

Authors

Contributions

R.M.M. performed the differentiation experiments. Q.L. carried out single-cell library preparations and S.A.S. analyzed single-cell data. O.R. and J.R.H. conducted and analyzed electrophysiological experiments. R.M.M., Y.M., and R.J.L. carried out all other experiments and data analyses. R.M.M. and S.P.P. conceived the project, designed experiments, and wrote the manuscript with input from all authors. S.P.P. supervised the work.

Corresponding author

Correspondence to Sergiu P. Pașca.

Ethics declarations

Competing interests

Stanford University has filed a provisional patent application that covers the generation of myelinating oligospheroids for studying human development and disease (US patent application number 15/953,197).

Additional information

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

Integrated supplementary information

Supplementary Figure 1 Characterization of oligodendrocytes, astrocytes, and neurons in hOLS.

a, Relative gene expression (normalized to GAPDH) as determined by qPCR at day 25 of in vitro culture of markers for pluripotency and germ layers (OCT4, SOX2, BRACH, SOX17) (n = 4 RNA samples of 2 hOLS each from 4 hiPS cell lines; see Supplementary Table 1; Kruskal–Wallis test, P = 0.001). b, Relative gene expression (normalized to GAPDH) in hCS and hOLS as determined by qPCR at day 37 of in vitro culture hOLS of CNS markers (FOXG1, SIX3, NKX2.1, OTX2, LMX1B, RAX1, HOXB4, PAX6, and LHX2) (n = 5 samples of 2 hOLS each from 5 hiPS cell lines; see Supplementary Table 1; Kruskal–Wallis test, P = 0.0001). c, Day 51 immunostaining of hOLS cryosections for OLIG2 and NKX2-2. Immunostainings were repeated on hOLS from four independent inductions with similar results. d, Histogram of the spatial distribution of MBP+ cells in hOLS cryosections. The number of MBP+ cells at various proportions of the radius (distance from center to edge) is shown. e, Relative gene expression (normalized to GAPDH) as determined by qPCR at day 100 of in vitro culture in hCS and hOLS of RBFOX3 (NEUN) (two-tailed t-test, t = 1.23, d.f. = 15, P = 0.23) and GFAP (two-tailed Mann–Whitney test, **P = 0.001) (for hCS n = 8 and for hOLS n = 9 RNA samples from spheroids derived from 4 hiPS cell lines in 1–4 differentiation experiments; see Supplementary Table 1). fi, Immunostaining of MAP2 at day 54 (f) and GFAP at day 110 (h) and quantification of MAP2+ (g) (two-tailed t-test, t = 3.82, d.f. = 7, **P = 0.006) and GFAP+ (i) (two-tailed t-test, t = 4.615, d.f. = 7, **P = 0.002) at day 54 and day 110 in dissociated hOLS (n = 5 samples each consisting of 4–6 hOLS derived from 3 hiPS cell lines; hiPS cell lines shown in different colors; see Supplementary Table 1). j, Relative expression of VGLUT1 (SLC17A7) (two-tailed Mann–Whitney test, ***P <0.0003) and GAD1 (two-tailed t-test, t = 2.92, d.f. = 17, **P = 0.009), in hCS and hOLS (normalized to GAPDH), as determined by qPCR at day 100 of in vitro culture (for hCS n = 8 and for hOLS n = 11 RNA samples from spheroids derived from 7 hiPS cell lines in 1–3 differentiation experiments; see Supplementary Table 1). k, Day 100 immunostaining of hOLS cryosections for GABA and MAP2. Immunostainings were repeated on hOLS from six independent inductions with similar results. Data are mean ± s.e.m. Scale bars, 50 μm (c, f, h, k upper panel), and 10 μm (k lower panel).

Supplementary Figure 2 Single cell characterization of hOLS.

a, Source of cells in the cluster in Fig. 2b. b, Single cell gene expression pattern of the markers for astrocytes (SOX9), endothelial cells (FLT1), neurons (STMN2), and myeloid cells (CX3CR1). c, Unsupervised hierarchical clustering of all single cells, colored by source. d, Relative gene expression (normalized by GAPDH) as determined by qPCR of MKI67, TOP2A, PDGFRA, and MBP in pooled cDNA from 50 single cells from the proliferating cells, OPCs, NFOs, and myelinating oligodendrocytes clusters (Fig. 2e-g). e, Percentage of cells in the proliferating cells, OPCs, NFOs, and myelinating oligodendrocytes subclusters in two hiPS cell lines. f, Pearson correlation values between the log normalized gene expression data for each oligodendrocyte subcluster between two hiPS cell lines.

Supplementary Figure 3 Primary and hOLS-derived oligodendrocyte differential gene expression and pattern of expression of disease-related genes.

a, Genes that were enriched in oligodendrocyte lineage cells isolated from primary tissue versus hOLS. b, Expression of the housekeeping gene GAPDH across pseudotime (colored by tissue of origin; log2 data normalized by gene). c, Single cell gene expression pattern of disease-implicated genes ARSA, RNASEH2A, and GALC in hOLS, hCS, and primary samples.

Supplementary Figure 4 Electrophysiological characterization of hOLS and myelination.

a, Quantification of the capacitance in bipolar and multipolar Sox10-MCS5::eGFP+ cells (n = 12 bipolar cells, n = 13 multipolar cells, two-tailed Mann–Whitney test, ***P = 0.0006). b, Quantification of the input resistance in bipolar and multipolar Sox10-MCS5::eGFP+ cells (n = 12 bipolar cells, n = 13 multipolar cells, two-tailed Mann–Whitney test, *P = 0.01). For a and b, dots represent individual cells, box edges represent s.e.m., the middle horizontal lines within the box represent the mean, and whiskers represent the 10th and 90th percentiles of the population. c, (upper left) Voltage clamp recording of a Sox10-MCS5::eGFP+ cell showing lack of inward current generation following electrical stimulation indicated by the red dot. (upper right) Voltage clamp recording of Sox10-MCS5::eGFP+ cell showing holding current variance in response to TTX (1 μM) and after treatment with NBQX and APC (lower right). Recordings were repeated in five cells from two independent inductions with similar results. d, Example images of interactions between MBP+ cells and NF-H+ processes in cryosections at days 150–158 of in vitro culture imaged by confocal microscopy. The first and seconds panels of each row are maximum projections, the third panel of each row is an individual z-section, and the right most panels are cross-sections. Immunostainings were repeated on hOLS from three independent inductions with similar results. e,f, Transmission electron microscopy images of hOLS from 8858-3 (e) and 0524-1 (f) at days 150–170 of differentiation showing stages of myelination. Electron microscopy was repeated on hOLS from three independent inductions with similar results. Scale bars, 1 μm (e, f), 50 μm (d left panel), and 10 μm (d middle panel).

Supplementary information

Supplementary Text and Figures

Supplementary Figs. 1–4 and Supplementary Tables 1–3.

Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Marton, R.M., Miura, Y., Sloan, S.A. et al. Differentiation and maturation of oligodendrocytes in human three-dimensional neural cultures. Nat Neurosci 22, 484–491 (2019). https://doi.org/10.1038/s41593-018-0316-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41593-018-0316-9

This article is cited by

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