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.

Cell diversity and network dynamics in photosensitive human brain organoids

Abstract

In vitro models of the developing brain such as three-dimensional brain organoids offer an unprecedented opportunity to study aspects of human brain development and disease. However, the cells generated within organoids and the extent to which they recapitulate the regional complexity, cellular diversity and circuit functionality of the brain remain undefined. Here we analyse gene expression in over 80,000 individual cells isolated from 31 human brain organoids. We find that organoids can generate a broad diversity of cells, which are related to endogenous classes, including cells from the cerebral cortex and the retina. Organoids could be developed over extended periods (more than 9 months), allowing for the establishment of relatively mature features, including the formation of dendritic spines and spontaneously active neuronal networks. Finally, neuronal activity within organoids could be controlled using light stimulation of photosensitive cells, which may offer a way to probe the functionality of human neuronal circuits using physiological sensory stimuli.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Figure 1: Large-scale, single-cell sequencing demonstrates development of a broad spectrum of cell types in human brain organoids.
Figure 2: Human brain organoids contain subclasses of forebrain and retinal cells.
Figure 3: Extended culture allows development of mature cell types including differentiated photoreceptors.
Figure 4: Brain organoids develop spontaneous networks and photosensitive neurons that can be modulated by sensory stimulation with light.

Accession codes

Primary accessions

Gene Expression Omnibus

References

  1. Kelava, I. & Lancaster, M. A. Stem cell models of human brain development. Cell Stem Cell 18, 736–748 (2016)

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  ADS  Google Scholar 

  3. Cugola, F. R. et al. The Brazilian Zika virus strain causes birth defects in experimental models. Nature 534, 267–271 (2016)

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  4. Dang, J. et al. Zika virus depletes neural progenitors in human cerebral organoids through activation of the innate immune receptor TLR3. Cell Stem Cell 19, 258–265 (2016)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Garcez, P. P. et al. Zika virus impairs growth in human neurospheres and brain organoids. Science 352, 816–818 (2016)

    Article  CAS  PubMed  ADS  Google Scholar 

  6. Nowakowski, T. J. et al. Expression analysis highlights AXL as a candidate Zika virus entry receptor in neural stem cells. Cell Stem Cell 18, 591–596 (2016)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. 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 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Quadrato, G., Brown, J. & Arlotta, P. The promises and challenges of human brain organoids as models of neuropsychiatric disease. Nat. Med. 22, 1220–1228 (2016)

    Article  CAS  PubMed  Google Scholar 

  10. Lancaster, M. A. & Knoblich, J. A. Generation of cerebral organoids from human pluripotent stem cells. Nat. Protocols 9, 2329–2340 (2014)

    Article  CAS  PubMed  Google Scholar 

  11. Macosko, E. Z. et al. Highly parallel genome-wide expression profiling of individual cells using nanoliter droplets. Cell 161, 1202–1214 (2015)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Satija, R., Farrell, J. A., Gennert, D., Schier, A. F. & Regev, A. Spatial reconstruction of single-cell gene expression data. Nat. Biotechnol. 33, 495–502 (2015)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Amir, A. D. et al. viSNE enables visualization of high dimensional single-cell data and reveals phenotypic heterogeneity of leukemia. Nat. Biotechnol. 31, 545–552 (2013)

    Article  CAS  PubMed Central  Google Scholar 

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

    MATH  Google Scholar 

  15. Arlotta, P. et al. Neuronal subtype-specific genes that control corticospinal motor neuron development in vivo. Neuron 45, 207–221 (2005)

    Article  CAS  PubMed  Google Scholar 

  16. Cahoy, J. D. et al. A transcriptome database for astrocytes, neurons, and oligodendrocytes: a new resource for understanding brain development and function. J. Neurosci. 28, 264–278 (2008)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Camp, J. G. et al. Human cerebral organoids recapitulate gene expression programs of fetal neocortex development. Proc. Natl Acad. Sci. USA 112, 15672–15677 (2015)

    Article  CAS  PubMed  ADS  PubMed Central  Google Scholar 

  18. 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  PubMed  ADS  PubMed Central  Google Scholar 

  19. Lake, B. B. et al. Neuronal subtypes and diversity revealed by single-nucleus RNA sequencing of the human brain. Science 352, 1586–1590 (2016)

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  20. Marques, S. et al. Oligodendrocyte heterogeneity in the mouse juvenile and adult central nervous system. Science 352, 1326–1329 (2016)

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  21. Molyneaux, B. J. et al. Novel subtype-specific genes identify distinct subpopulations of callosal projection neurons. J. Neurosci. 29, 12343–12354 (2009)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Pollen, A. A. et al. Molecular identity of human outer radial glia during cortical development. Cell 163, 55–67 (2015)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Siegert, S. et al. Transcriptional code and disease map for adult retinal cell types. Nat. Neurosci. 15, 487–495 (2012)

    Article  CAS  PubMed  Google Scholar 

  24. Strunnikova, N. V. et al. Transcriptome analysis and molecular signature of human retinal pigment epithelium. Hum. Mol. Genet. 19, 2468–2486 (2010)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Xia, N. et al. Transcriptional comparison of human induced and primary midbrain dopaminergic neurons. Sci. Rep. 6, 20270 (2016)

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  26. Zhang, Y. et al. An RNA-sequencing transcriptome and splicing database of glia, neurons, and vascular cells of the cerebral cortex. J. Neurosci. 34, 11929–11947 (2014)

    Article  CAS  PubMed  PubMed Central  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  PubMed  Google Scholar 

  28. Furukawa, T., Morrow, E. M. & Cepko, C. L. Crx, a novel otx-like homeobox gene, shows photoreceptor-specific expression and regulates photoreceptor differentiation. Cell 91, 531–541 (1997)

    Article  CAS  PubMed  Google Scholar 

  29. Korf, H. W., White, B. H., Schaad, N. C. & Klein, D. C. Recoverin in pineal organs and retinae of various vertebrate species including man. Brain Res. 595, 57–66 (1992)

    Article  CAS  PubMed  Google Scholar 

  30. Dennis, G. Jr et al. DAVID: database for annotation, visualization, and integrated discovery. Genome Biol. 4, 3 (2003)

    Article  Google Scholar 

  31. Huttenlocher, P. R. & Dabholkar, A. S. Regional differences in synaptogenesis in human cerebral cortex. J. Comp. Neurol. 387, 167–178 (1997)

    Article  CAS  PubMed  Google Scholar 

  32. Scholvin, J. et al. Close-packed silicon microelectrodes for scalable spatially oversampled neural recording. IEEE Trans. Biomed. Eng. 63, 120–130 (2016)

    Article  PubMed  PubMed Central  Google Scholar 

  33. Fujisawa, S., Amarasingham, A., Harrison, M. T. & Buzsáki, G. Behavior-dependent short-term assembly dynamics in the medial prefrontal cortex. Nat. Neurosci. 11, 823–833 (2008)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Stark, E. & Abeles, M. Unbiased estimation of precise temporal correlations between spike trains. J. Neurosci. Methods 179, 90–100 (2009)

    Article  PubMed  Google Scholar 

  35. Zeman, S. & Lodge, D. Pharmacological characterization of non-NMDA subtypes of glutamate receptor in the neonatal rat hemisected spinal cord in vitro. Br. J. Pharmacol. 106, 367–372 (1992)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Zhong, X. et al. Generation of three-dimensional retinal tissue with functional photoreceptors from human iPSCs. Nat. Commun. 5, 4047 (2014)

    Article  CAS  PubMed  ADS  Google Scholar 

  37. Boulting, G. L. et al. A functionally characterized test set of human induced pluripotent stem cells. Nat. Biotechnol. 29, 279–286 (2011)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Quadrato, G. et al. Nuclear factor of activated T cells (NFATc4) is required for BDNF-dependent survival of adult-born neurons and spatial memory formation in the hippocampus. Proc. Natl Acad. Sci. USA 109, E1499–E1508 (2012)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Quadrato, G. et al. Long-term culture and electrophysiological characterization of human brain organoids. Protoc. Exch. http://dx.doi.org/10.1038/protex.2017.049 (2017)

  40. Waltman, L. & van Eck, N. J. A smart local moving algorithm for large-scale modularity-based community detection. Eur. Phys. J. B 86, 471 (2013)

    Article  ADS  CAS  Google Scholar 

  41. Chung, N. C. & Storey, J. D. Statistical significance of variables driving systematic variation in high-dimensional data. Bioinformatics 31, 545–554 (2015)

    Article  CAS  PubMed  Google Scholar 

  42. Kasthuri, N. et al. Saturated reconstruction of a volume of neocortex. Cell 162, 648–661 (2015)

    Article  CAS  PubMed  Google Scholar 

  43. Rossant, C. et al. Spike sorting for large, dense electrode arrays. Nat. Neurosci. 19, 634–641 (2016)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Eden, U. T. & Kramer, M. A. Drawing inferences from Fano factor calculations. J. Neurosci. Methods 190, 149–152 (2010)

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

We thank J.R. Brown, S. Hyman, G. Feng, Z. Fu, A. Schinder, L. Rubin, F. Rapino, former and present members of the Arlotta laboratory for insightful discussions and editing of the manuscript, A. Pollen and A. Kriegstein for sharing of human single-cell datasets and C. Cepko for sharing of antibodies, Y. Zhang for outstanding technical support, E. Zuccaro, F. Yates and S. Pavoni for helpful advice on culturing organoids. This work was supported by grants from the Stanley Center for Psychiatric Research, the Broad Institute of Harvard and MIT, and the Star Family Award of Harvard University to P.A. E.S.B. acknowledges NIH Director's Pioneer Award 1DP1NS087724. J.W.L. acknowledges support by IARPA, Conte and MURI Army Research Office. P.A. and E.S.B. are New York Stem Cell Foundation-Robertson Investigators.

Author information

Authors and Affiliations

Authors

Contributions

P.A. and G.Q. conceived the experiments. G.Q. developed the long-term cultures of organoids and performed all immunohistochemical characterization with help from N.M. E.Z.M., G.Q., T.N. and M.G. performed all single-cell sequencing experiments. E.Z.M., G.Q., T.N., P.A. and S.A.M. analysed and interpreted the Drop-seq data. J.L.S., S.M.Y. and Z.M.W. performed electrophysiological experiments. J.S., J.P.K. and E.S.B. developed multi-electrode probes and helped J.L.S. adapt them to organoids. D.R.B. and J.W.L. performed electron microscopy work. P.A., G.Q., E.Z.M. and S.A.M. wrote the manuscript with contributions from all authors.

Corresponding authors

Correspondence to Giorgia Quadrato or Paola Arlotta.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Additional information

Reviewer Information Nature thanks F. Guillemot, S. Linnarsson and the other anonymous reviewer(s) for their contribution to the peer review of this work.

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

Extended data figures and tables

Extended Data Figure 1 Time-course of expression of selected marker genes in human brain organoids.

a, Top, expression of the hypoxia marker HIF1α over one to nine months of culture, and positive control (a six-month-old organoid treated with cobalt (II) chloride (CoCl), an activator of the hypoxia signalling pathway). Bottom, expression of the apoptosis marker, active caspase 3 (ActCasp3) and the neuronal marker (MAP2), over one to nine months of culture. b, Expression of markers for progenitor, neuronal and glial populations over one to nine months of culture. c, One-month-old brain organoids exhibit early brain regionalization, expressing markers of forebrain (PAX6 and NKX2.1), midbrain (OTX2) and hindbrain (GBX2) progenitors. d, One-month-old brain organoids express the cortical marker EMX1, the forebrain marker FOXG1 and the retina marker VSX2, with spatial segregation between regions positive for forebrain versus retinal markers. Scale bars, 250 μm and 20 μm (insets).

Extended Data Figure 2 Decoding of the identity of cell types within the main clusters.

a, Heatmap of differentially expressed cell-type marker genes across each of the 10 main clusters. For gene lists and P values, see Supplementary Table 2, for references of cluster identification, see Extended Data Table. 1. b, Heatmap of average expression for representative marker genes and cell-type classification of the main clusters from the six-month-old organoid dataset.

Extended Data Figure 3 Quantifying variability among organoids.

tSNE plots depicting the proportion of cells each organoid contributed to each cell-type cluster. The cells are colour-coded by the organoid of origin. The large plot shows the entire cluster, and the smaller plots show the contribution of each individual organoid, labelled with the percentage of cells within the cluster contributed by that organoid.

Extended Data Figure 4 Correlation analysis for organoid cell types compared to the human fetal cortex and mouse retina.

a, Correlation between expression patterns of highly variable genes between cell populations within a previously published single-cell RNA-seq dataset of the human fetal cortex22 against the organoid astroglial cluster (c2) and the identified subclusters of the forebrain cluster (c4). IPC, intermediate progenitor cells. b, Correlation between expression patterns of highly variable genes between cell populations within a previously published single-cell RNA-seq dataset of postnatal day (P)14 mouse retina11 and the cell populations identified within the retinal cluster (c5) in our organoid dataset. Pigmented Epi, pigmented epithelium; RGC, retinal ganglion cells. Colour keys (and dot sizes) represent the range of the coefficients of determination (r2) in each analysis.

Extended Data Figure 5 Distribution and reproducibility of forebrain and retinal cell types across organoids.

a, Immunohistochemical detection in a six-month-old organoid of the forebrain marker FOXG1 with the progenitor markers Nestin and SOX2 (left) and of FOXG1 with the corticofugal projection neuron marker CTIP2 and the callosal projection neuron marker SATB2 (right), all showing extensive colocalized coexpression, which was observed in all organoids examined (6 out of 6). b, Number of cells assigned to each cortical cell type in individual organoids from flask 3, and the percentage of each cell type out of all classified cortical cells in that organoid. c, Immunohistochemical detection of retinal cell types in six-month-old organoids shows expression of known markers for amacrine cells, bipolar cells, retinal ganglion cells, Müller glia, rods and retinal pigmented epithelium, observed across all organoids in every flask, n = 11; 3 bioreactors. d, Number of cells assigned to each retinal cell type in individual organoids from flasks 1–3, and the percentage of each cell type out of all classified retinal cells in that organoid. AC, amacrine cells; BP, bipolar cells; CfuPN, corticofugal projection neurons; CPN, callosal projection neurons; IN, interneurons; IPC, intermediate progenitor cells; MG, Müller glia; PE, pigmented epithelium; photo, photoreceptors; RG, radial glial cells; RGC, retinal ganglion cells.

Extended Data Figure 6 Electron microscopy of an eight-month-old human brain organoid.

a, Whole organoid before vibratome sectioning shows regional structures. The red line shows the approximate location of the vibratome slice. b, Example 40-nm section. c. Regular finger-like membrane stack could indicate development of the outer segment of a retinal rod cell. d, Stacked endoplasmic reticulum. e, Example synapse with crystalline-looking arrangement of vesicles. f, Putative muscle cell characterized by a large cell body with a diameter of about 30 μm highlighted in an organoid section stained with eosin and haematoxylin. g, Example of a muscle cell showing a distribution of the mitochondria near the cell surface, which is typical for muscle cells. h, Several cell bodies with different morphologies and cytoplasm content highlighted in colour. i, Large cell body with muscle fibres highlighted in blue. Inset (red square) shows muscle fibres with sarcomeres. j, One of the dark cell bodies appearing as dark spots on the organoid surface. k, Cell reaching to the surface of the organoid. Scale bars, 50 μm (f), 10 μm (gk) and 1 μm (i, inset).

Extended Data Figure 7 Statistical structure of spontaneous activity.

a, Electron micrographs of a 64-channel probe shank tip (Fig. 4a). Scale bars, 100 μm (top) and 20 μm (bottom). b, Example inter-spike interval (ISI, top) and auto-correlogram (bottom) plots for spontaneous activity recorded from 4 prototypical units; a.u., arbitrary units; freq., frequency. c, Plot of the mean spike rate in 11a cells (dots) and HuES66 (+) organoids recorded at 7–9 months. The difference between the median of the mean spike rates is not significant across lines (11a cells, n = 34 cells, 12 recording sites, 7 organoids, median = 0.662 Hz, Q1–Q3 = 0.186–2.071 Hz; HuES66 n = 27 cells, 9 recording sites, 8 organoids, median = 1.186 Hz, Q1–Q3= 0.463–2.848 Hz; two-tailed Wilcoxon rank-sum test, z = –1.47, P = 0.14; furthermore, the variance is not significantly different, squared-ranks test, z = –1.47, P = 0.141). d, Plot of the Fano factor against organoid age. e, Plot of mean spike rate against the Fano factor (n = 61 neuronal units, from 15 organoids). Fano factors outside the expected 99% confidence bounds for a Poisson distribution are plotted in grey (otherwise black). f, Plot of the Fano factors calculated across the whole recording versus the first second of population bursts. Shading, 99% confidence bounds for a whole-recording Fano factor of 1. Black symbols, Fano factor close to 1, consistent with a stationary Poisson-distributed system. g, Spike train auto-correlogram (colour-coded as in Fig. 4g) and cross-correlogram (grey) for the three units presented in Fig. 4g. h, Left, baseline normalized spike rate (n = 25 cells, 10 organoids; 11a cells, n = 9 cells, 4 organoids; HuES66, n = 16 cells, 6 organoids); the clustering of data of responders (red) and non-responders (blue). Middle, count histogram of the left data plot overlaid by the optimal Gaussian mixture model (number of components k = 2, see methods; μk = kth mean, σk = kth s.d.). The underlying distributions are shaded (μ1 = 0.16, σ1 = 0.11, mixing proportion = 0.16; μ2 = 0.99, σ2 = 0.18, mixing proportion = 0.84). Right, plot of the Bayes information criterion (BIC) generated for the Gaussian mixture models with k components. i, Population averaged activity for light responsive (red, n = 4), and non-responsive (blue, n = 21), neurons; error envelope (shaded) is the s.e.m. In the four responsive organoids, light stimulation attenuated firing rate in 4 out of 5 isolated neurons.

Extended Data Table 1 References used for cluster identification in Extended Data Fig. 2

Supplementary information

Supplementary Information

This file contains a Supplementary Discussion and Supplementary Table 1, a list of primary antibodies employed in this study. (PDF 244 kb)

Supplementary Table 2

Genes differentially expressed in each of the 10 main clusters relative to the rest of the data set (first tab), and differential gene expression between each subcluster (remaining tabs). Bracketed numbers at the head of each section designate the cluster or subcluster. Column headings: p_val: p-value significance of expression level; avg_diff: log average differential expression between groups; pct.1: Fraction of cells expressing gene in group 1; pct.2: Fraction of cells expressing gene in group 2 (comparison group). (XLSX 752 kb)

Slice animation of the aligned EM image stack with reconstructed objects as color overlay

Organoid surface towards the top of the images. (MP4 27916 kb)

Reconstructed axons and dendrites in the same volume as video 1, shown as 3D objects

Organoid surface towards the top. (MP4 17598 kb)

Three spine synapses between two axons and two dendrites, two made en-passant and one on a terminal bouton

Same as Figure 3j, with red dendrite added. (MP4 3795 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Quadrato, G., Nguyen, T., Macosko, E. et al. Cell diversity and network dynamics in photosensitive human brain organoids. Nature 545, 48–53 (2017). https://doi.org/10.1038/nature22047

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature22047

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