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:

Generation of ‘semi-guided’ cortical organoids with complex neural oscillations

Abstract

Temporal development of neural electrophysiology follows genetic programming, similar to cellular maturation and organization during development. The emergent properties of this electrophysiological development, namely neural oscillations, can be used to characterize brain development. Recently, we utilized the innate programming encoded in the human genome to generate functionally mature cortical organoids. In brief, stem cells are suspended in culture via continuous shaking and naturally aggregate into embryoid bodies before being exposed to media formulations for neural induction, differentiation and maturation. The specific culture format, media composition and duration of exposure to these media distinguish organoid protocols and determine whether a protocol is guided or unguided toward specific neural fate. The ‘semi-guided’ protocol presented here has shorter induction and differentiation steps with less-specific patterning molecules than most guided protocols but maintains the use of neurotrophic factors such as brain-derived growth factor and neurotrophin-3, unlike unguided approaches. This approach yields the cell type diversity of unguided approaches while maintaining reproducibility for disease modeling. Importantly, we characterized the electrophysiology of these organoids and found that they recapitulate the maturation of neural oscillations observed in the developing human brain, a feature not shown with other approaches. This protocol represents the potential first steps toward bridging molecular and cellular biology to human cognition, and it has already been used to discover underlying features of human brain development, evolution and neurological conditions. Experienced cell culture technicians can expect the protocol to take 1 month, with extended maturation, electrophysiology recording, and adeno-associated virus transduction procedure options.

Key points

  • This protocol describes the generation of cortical organoids with complex neural oscillations using a ‘semi-guided’ approach, as well as their functional characterization through microelectrode array measurements, calcium imaging and adeno-associated virus transduction.

  • The ‘semi-guided’ nature of this approach allows an intermediate between the cellular heterogeneity of unguided approaches and the predictability of guided approaches, which we speculate underlies the emergence of complex oscillations.

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: Overview of generation of cortical organoids and resulting features.
Fig. 2: Electrophysiology overview and characterization.
Fig. 3: Calcium imaging overview and AAV7m8–GCaMP transduction characterization in cortical organoids.

Similar content being viewed by others

Data availability

Further information for Figs. 1d,e and 2c–f can be found in Trujillo et al.7. Sequencing from Trujillo et al. for Fig. 1 can be found at National Center for Biotechnology Information Gene Expression Omnibus: GSE130238. Further information for Extended Data Fig. 3d–f can be found in Tanaka et al.8.

Code availability

Code for MEA processing can be found at https://github.com/voytekresearch/OscillatoryOrganoids/blob/master/organoid_EEG_age_regression.ipynb.

References

  1. Scuderi, S., Altobelli, G., Cimini, V., Coppola, G. & Vaccarino, F. Cell-to-cell adhesion and neurogenesis in human cortical development: a study comparing 2D monolayers with 3D organoid cultures. Stem Cell Rep. 16, 264–280 (2021).

    Article  CAS  Google Scholar 

  2. Centeno, E., Cimarosti, H. & Bithell, A. 2D versus 3D human induced pluripotent stem cell-derived cultures for neurodegenerative disease modelling. Mol. Neurodegener. 13, 27 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  3. Adlakha, Y. Human 3D brain organoids: steering the demolecularization of brain and neurological diseases. Cell Death Discov. 9, 221 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Grenier, K., Kao, J. & Diamandis, P. Three-dimensional modeling of human neurodegeneration: brain organoids coming of age. Mol. Psychiatry 25, 254–274 (2020).

    Article  PubMed  Google Scholar 

  5. 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 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Trujillo, C. et al. Complex oscillatory waves emerging from cortical organoids model early human brain network development. Cell. Stem Cell. 25, 558–569.e7 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Tanaka, Y., Cakir, B., Xiang, Y., Sullivan, G. & Park, I. Synthetic analyses of single-cell transcriptome from multiple brain organoids and fetal brain. Cell Rep. 30, 1682–1689 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Trujillo, C. et al. Reintroduction of the archaic variant of NOVA1 in cortical organoids alters neurodevelopment. Science 371, eaax2537 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Papes, F. et al. Transcription factor 4 loss-of-function is associated with deficits in progenitor proliferation and cortical neuron content. Nat. Commun. 13, 2387 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Allison, T. et al. Defining the nature of human pluripotent stem cell-derived interneurons via single-cell analysis. Stem Cell Rep. 16, 2548–2564 (2021).

    Article  CAS  Google Scholar 

  12. Delgado, R. et al. Individual human cortical progenitors can produce excitatory and inhibitory neurons. Nature 601, 397–403 (2022).

    Article  CAS  PubMed  Google Scholar 

  13. Zourray, C., Kurian, M., Barral, S. & Lignani, G. Electrophysiological properties of human cortical organoids: current state of the art and future directions. Front. Mol. Neurosci. 15, 839366 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Xiang, Y. et al. Fusion of regionally-specified hPSC-derived organoids models human brain development and interneuron migration. Cell Stem Cell 21, 383–398.e7 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Fiddes, I. et al. Human-specific NOTCH2NL genes affect notch signaling and cortical neurogenesis. Cell 173, 1356–1369.e22 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Madhavan, M. et al. Induction of myelinating oligodendrocytes in human cortical spheroids. Nat. Methods 15, 700–706 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  20. Quadrato, G. et al. Cell diversity and network dynamics in photosensitive human brain organoids. Nature 545, 48–53 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Sharf, T. et al. Functional neuronal circuitry and oscillatory dynamics in human brain organoids. Nat. Commun. 13, 4403 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Uhlhaas, P., Roux, F., Rodriguez, E., Rotarska-Jagiela, A. & Singer, W. Neural synchrony and the development of cortical networks. Trends Cogn. Sci. 14, 72–80 (2010).

    Article  PubMed  Google Scholar 

  23. de Hemptinne, C. et al. Therapeutic deep brain stimulation reduces cortical phase-amplitude coupling in Parkinson’s disease. Nat. Neurosci. 18, 779–786 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Uhlhaas, P. J. & Singer, W. Abnormal neural oscillations and synchrony in schizophrenia. Nat. Rev. Neurosci. 11, 100–113 (2010).

    Article  CAS  PubMed  Google Scholar 

  25. Khan, S. et al. Local and long-range functional connectivity is reduced in concert in autism spectrum disorders. Proc. Natl Acad. Sci. USA 110, 3107–3112 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Dixon, T. & Muotri, A. Advancing preclinical models of psychiatric disorders with human brain organoid cultures. Mol. Psychiatry 28, 83–95 (2023).

    Article  PubMed  Google Scholar 

  27. Samarasinghe, R. et al. Identification of neural oscillations and epileptiform changes in human brain organoids. Nat. Neurosci. 24, 1488–1500 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Lavazza, A. ‘Consciousnessoids’: clues and insights from human cerebral organoids for the study of consciousness. Neurosci. Conscious. 7, niab029 (2021).

    Article  PubMed  Google Scholar 

  29. Adams, J. et al. Impact of alcohol exposure on neural development and network formation in human cortical organoids. Mol. Psychiatry 28, 1571–1584 (2023).

    Article  CAS  PubMed  Google Scholar 

  30. Mesci, P. et al. Modeling neuro–immune interactions during Zika virus infection. Hum. Mol. Genet. 27, 41–52 (2018).

    Article  CAS  PubMed  Google Scholar 

  31. Schley, L. Meet the scientists connecting lab-grown ‘mini brains’ to robots. Discover Magazine https://www.discovermagazine.com/mind/meet-the-scientists-connecting-lab-grown-mini-brains-to-robots (2019).

  32. Marinho, L. et al. The impact of antidepressants on human neurodevelopment: brain organoids as experimental tools. Semin. Cell Dev. Biol. 144, 67–76 (2023).

    Article  CAS  PubMed  Google Scholar 

  33. Trujillo, C. et al. Pharmacological reversal of synaptic and network pathology in human MECP2-KO neurons and cortical organoids. EMBO Mol. Med. 13, e12523 (2021).

    Article  CAS  PubMed  Google Scholar 

  34. Adams, J. W., Cugola, F. R. & Muotri, A. R. Brain organoids as tools for modeling human neurodevelopmental disorders. Physiology 34, 365–375 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Coelho, L. & Muotri, A. Cortical brain organoid as a model to study microgravity exposure. Artif. Organs 47, 5–7 (2023).

    Article  PubMed  Google Scholar 

  36. Standards document. International Society for Stem Cell Research https://www.isscr.org/standards-document (2023).

  37. Lin, M. & Schnitzer, M. Genetically encoded indicators of neuronal activity. Nat. Neurosci. 19, 1142–1153 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  38. Avansini, S. et al. Junctional instability in neuroepithelium and network hyperexcitability in a focal cortical dysplasia human model. Brain 145, 1962–1977 (2022).

    Article  PubMed  Google Scholar 

  39. Dalkara, D. et al. In vivo-directed evolution of a new adeno-associated virus for therapeutic outer retinal gene delivery from the vitreous. Sci. Transl. Med. 5, 189ra76 (2013).

    Article  PubMed  Google Scholar 

  40. Duong, T. et al. Comparative AAV–eGFP transgene expression using vector serotypes 1–9, 7m8, and 8b in human pluripotent stem cells, RPEs, and human and rat cortical neurons. Stem Cells Int. 2019, 7281912 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  41. Xu, D. et al. Overexpressing NeuroD1 reprograms Müller cells into various types of retinal neurons. Neural Regen. Res. 18, 1124–1131 (2023).

    Article  CAS  PubMed  Google Scholar 

  42. Garita-Hernandez, M. et al. AAV-mediated gene delivery to 3D retinal organoids derived from human induced pluripotent stem cells. Int. J. Mol. Sci. 21, 994 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. McClements, M. et al. Tropism of AAV vectors in photoreceptor-like cells of human iPSC-derived retinal organoids. Transl. Vis. Sci. Technol. 11, 3 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  44. Zarowny, L. et al. Bright and high-performance genetically encoded Ca2+ indicator based on mneongreen fluorescent protein. ACS Sens. 5, 1959–1968 (2020).

    Article  CAS  PubMed  Google Scholar 

  45. Piedra, J. et al. Development of a rapid, robust, and universal picogreen-based method to titer adeno-associated vectors. Hum. Gene Ther. Methods 26, 35–42 (2015).

    Article  CAS  PubMed  Google Scholar 

  46. Puppo, F. et al. All-optical electrophysiology in hiPSC-derived neurons with synthetic voltage sensors. Front. Cell Neurosci. 15, 671549 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Mukamel, E., Nimmerjahn, A. & Schnitzer, M. Automated analysis of cellular signals from large-scale calcium imaging data. Neuron 63, 747–760 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Giovannucci, A. et al. CaImAn an open source tool for scalable calcium imaging data analysis. eLife 8, e3817343 (2019).

    Article  Google Scholar 

  49. Gordon, A. et al. Long-term maturation of human cortical organoids matches key early postnatal transitions. Nat. Neurosci. 24, 331–342 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Giandomenico, S. et al. Cerebral organoids at the air-liquid interface generate diverse nerve tracts with functional output. Nat. Neurosci. 22, 669–679 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors thank I.-H. Park and colleagues for approving the inclusion of published data (Supplementary Fig. 3) adapted from their figure list in Tanaka et al.8. Additionally, we acknowledge the Muotri Lab for their discussion on best organoid culture practices. This work was supported by US National Institutes of Health grants (R01MH100175, R01NS105969, MH123828, R01NS123642, R01MH127077, R01ES033636, R21MH128827, R01AG078959, R01DA056908, R01HD107788, R01HG012351, R21HD109616, R01MH107367), a California Institute for Regenerative Medicine grant (DISC2-13515) and a grant from the Department of Defense (W81XWH2110306).

Author information

Authors and Affiliations

Authors

Contributions

M.Q.F., T.C. and A.R.M. updated existing protocol documents to match current best practices and devised a protocol manuscript outline. M.Q.F. wrote the first full draft of the protocol document, generated Figs. 1–2 and Extended Data Fig. 2 from existing laboratory documents and original additions, and formatted the document for submission. M.Q.F., T.C., F.P. and A.R.M. edited the manuscript and devised content for additional experimental procedures following the main protocol. M.Q.F. and T.C. wrote the comparison between protocols in the introduction and troubleshooting tips for organoid culture and quality control. T.C. wrote the AggreWell forced aggregation protocol in the Supplementary Methods, Procedure 2: MEA protocol, the MEA section of the ‘Anticipated results’ and Extended Data Fig. 3. F.P. wrote Procedure 3: calcium imaging, troubleshooting for Options B and C, and provided Supplementary Videos 1, 2 and 3. F.P. and R.B. wrote Procedure 4: AAV transduction, and provided Fig. 3. A.R.M., S.S. and M.C. oversaw generation of the protocol. All authors were involved in final editing of the protocol.

Corresponding author

Correspondence to Alysson R. Muotri.

Ethics declarations

Competing interests

A.R.M. is a cofounder of and has an equity interest in TISMOO, a company dedicated to genetic analysis and brain organoid modeling focusing on therapeutic applications customized for autism spectrum disorder and other neurological disorders with genetic origins. The terms of this arrangement have been reviewed and approved by the University of California San Diego in accordance with its conflict-of-interest policies. A.R.M. is an inventor of several patents related to human functional brain organogenesis, including the protocol described here.

Peer review

Peer review information

Nature Protocols thanks Wei Niu, Ranmal Samarasinghe, Tjitse Vandermolen and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

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

Related links

Key references using this protocol

Trujillo, C. et al. Cell Stem Cell. 25, 558–569.e7 (2019): https://doi.org/10.1016/j.stem.2019.08.002

Trujillo, C. et al. Science 371, eaax2537 (2021): https://doi.org/10.1126/science.aax2537

Papes, F. et al. Nat. Commun. 13, 2387 (2022): https://doi.org/10.1038/s41467-022-29942-w

Extended data

Extended Data Fig. 1 Third-party comparison of different brain organoid protocols and the fetal human brain.

(a) Schematic View of the Culture System Generating human cortical organoids (hCOs). Guided protocols originated from Eiraku et al.5 while non-guided protocols are from Lancaster et al.19. Timeline of neural induction, differentiation, and maturation step is shown across protocols. Note that, while we have use the term ‘semi-guided’ here to describe our protocol and distinguish it from other Eiraku et al.5 -derived directed protocols, all panels in this figure are adapted from ref. 8, which only utilized the terms ‘guided’ and ‘unguided’ and thus correctly classified our protocol7 as guided. (b–d) Shared-nearest-neighbors (SNN) graph visualization for differentiation trajectory. (b) Differentiation directions (arrows) were determined by pseudotime. (c) Estimated trajectory backbone from the SNN graph. (d) Comparison of differentiation trajectory among different protocols. (e) The presence of cell types in each organoid protocol and human fetal brain. Cell types with >0.25% of cells are denoted with a plus sign. F, Fiddes et al.17; V, Velasco et al.6; B, Birey et al.14; M, Madhavan et al.18; T, Trujillo et al.7; X, Xiang et al.15; Q, Quadrato et al.20; G, Giandomenico et al.50. (f) Enrichment of disease-related genes in each organoid protocol. The red boxes indicate data generated from the protocol presented here. Figure adapted with permission from ref. 8, Elsevier.

Extended Data Fig. 2 Molecular and functional reproducibility of cortical organoids.

(a) Schematic showing the single-cell approach performed to assess the reproducibility of organoid generation using different iPSC lines (WT1 and WT2). (b) tSNE plot of single-cell mRNA sequencing data from two 6-month-old organoids. (c) Expression of gene markers for various cell types both batches. (d) Population ratio of each cluster by replicate. (e) Consistent and reproducible development of electrical activity in organoids over time across four cell lines, bars represent mean ± s.e.m (n = 8, independent experiments performed in duplicates using two clones of an iPSC line).

Extended Data Fig. 3 Selection of organoids for plating.

(a) Good-quality 1-month-old organoids with visible spatial arrangement of neural rosette structures. Scale bar, 1,000 μm. Mixed quality organoids, where there is a mixed population of fully differentiated organoids with visible rosette structures and incomplete differentiated organoids—select rosetted organoids only for downstream assays and discard incompletely differentiated organoids. Poor-quality spheroids that did not efficiently neuralize and differentiate into rosetted organoids—discard and start over. (b) Distinguish proper spatial organization and structural development in organoids; orange arrows indicate holes and green arrows indicate rosettes. Scale bar, 1,000 μm. (c) Use of immunohistochemistry to distinguish between good-quality organoids with spatial neural rosette arrangement (smaller circles within the organoid) and incompletely differentiated organoids that contain neurons but lack neural rosette structures and spatial organization. Immunostainings showing nuclei (DAPI), neuron microtubules (MAP2), and proliferating NPCs (Ki67 and Nestin). Scale bar, 50 μm.

Supplementary information

Supplementary Information

Supplementary Methods.

Supplementary Video 1

Representative movies of calcium activity in GCaMP-expressing cortical organoids

Supplementary Video 2

Representative movies of calcium activity in OGB1-labeled 2D cultures of iPSC-derived neurons. 2D cortical neurons are 4 and 5 months old, respectively.

Supplementary Video 3

Representative movies of calcium activity from neurons from OGB1- labeled organoids plated on imaging plates.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Fitzgerald, M.Q., Chu, T., Puppo, F. et al. Generation of ‘semi-guided’ cortical organoids with complex neural oscillations. Nat Protoc (2024). https://doi.org/10.1038/s41596-024-00994-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41596-024-00994-0

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