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:

Brain Chimeroids reveal individual susceptibility to neurotoxic triggers

Abstract

Interindividual genetic variation affects the susceptibility to and progression of many diseases1,2. However, efforts to study how individual human brains differ in normal development and disease phenotypes are limited by the paucity of faithful cellular human models, and the difficulty of scaling current systems to represent multiple people. Here we present human brain Chimeroids, a highly reproducible, multidonor human brain cortical organoid model generated by the co-development of cells from a panel of individual donors in a single organoid. By reaggregating cells from multiple single-donor organoids at the neural stem cell or neural progenitor cell stage, we generate Chimeroids in which each donor produces all cell lineages of the cerebral cortex, even when using pluripotent stem cell lines with notable growth biases. We used Chimeroids to investigate interindividual variation in the susceptibility to neurotoxic triggers that exhibit high clinical phenotypic variability: ethanol and the antiepileptic drug valproic acid. Individual donors varied in both the penetrance of the effect on target cell types, and the molecular phenotype within each affected cell type. Our results suggest that human genetic background may be an important mediator of neurotoxin susceptibility and introduce Chimeroids as a scalable system for high-throughput investigation of interindividual variation in processes of brain development and disease.

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: NSC-Chimeroids maintain donor representation across cell classes.
Fig. 2: NSC- and NPC-Chimeroids control hyperproliferative states of starting PSCs.
Fig. 3: Chimeroids are reproducible and display appropriate cell type composition.
Fig. 4: Treatment-specific alterations in the NSC-Chimeroids model.
Fig. 5: NSC-Chimeroids reveal differential donor-specific susceptibility.

Similar content being viewed by others

Data availability

Read-level data from scRNA-seq have been deposited at Synapse (https://www.synapse.org/; syn52132869), while count-level data and metadata have been deposited at https://singlecell.broadinstitute.org/single_cell/study/SCP2609. Any additional information required to reanalyse the data reported in this paper is available from the corresponding author on request. Data from previous publications that were used in this study were as follows: organoid reference map and fetal data20 (Synapse: syn26346373); fetal data23 (GEO: GSE162170) and fetal data24 (dbGaP: phs001836).

Code availability

Code used during data analysis is available at GitHub (https://github.com/tfaits/Arlotta_Lab_Chimeroids).

References

  1. Paulsen, B. et al. Autism genes converge on asynchronous development of shared neuron classes. Nature 602, 268–273 (2022).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  2. Pizzo, L. et al. Rare variants in the genetic background modulate cognitive and developmental phenotypes in individuals carrying disease-associated variants. Genet. Med. 21, 816–825 (2019).

    Article  CAS  PubMed  Google Scholar 

  3. Ford, L. C. et al. A population-based human in vitro approach to quantify inter-individual variability in responses to chemical mixtures. Toxics 10, 441 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Germain, P.-L. & Testa, G. Taming human genetic variability: transcriptomic meta-analysis guides the experimental design and interpretation of iPSC-based disease modeling. Stem Cell Rep. 8, 1784–1796 (2017).

    Article  Google Scholar 

  5. Tegtmeyer, M. et al. High-dimensional phenotyping to define the genetic basis of cellular morphology. Nat. Commun. 15, 347 (2024).

  6. Cederquist, G. Y. et al. A multiplex human pluripotent stem cell platform defines molecular and functional subclasses of autism-related genes. Cell Stem Cell 27, 35–49 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Cuomo, A. S. E. et al. Single-cell RNA-sequencing of differentiating iPS cells reveals dynamic genetic effects on gene expression. Nat. Commun. 11, 810 (2020).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  8. Jerber, J. et al. Population-scale single-cell RNA-seq profiling across dopaminergic neuron differentiation. Nat. Genet. 53, 304–312 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Limone, F. et al. Efficient generation of lower induced motor neurons by coupling Ngn2 expression with developmental cues. Cell Rep. https://doi.org/10.1016/j.celrep.2022.111896 (2023).

  10. Mitchell, J. M. et al. Mapping genetic effects on cellular phenotypes with “cell villages”. Preprint at bioRxiv https://doi.org/10.1101/2020.06.29.174383 (2020).

  11. Wells, M. F. et al. Natural variation in gene expression and viral susceptibility revealed by neural progenitor cell villages. Cell Stem Cell 30, 312–332 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Wozniak, J. R., Riley, E. P. & Charness, M. E. Clinical presentation, diagnosis, and management of fetal alcohol spectrum disorder. Lancet Neurol. 18, 760–770 (2018).

    Article  Google Scholar 

  13. Bjørk, M.-H. et al. Association of prenatal exposure to antiseizure medication with risk of autism and intellectual disability. JAMA Neurol. 79, 672–681 (2022).

    Article  PubMed  Google Scholar 

  14. Christensen, J. et al. Prenatal valproate exposure and risk of autism spectrum disorders and childhood autism. JAMA 309, 1696–1703 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Neavin, D. R. et al. A village in a dish model system for population-scale hiPSC studies. Nat. Commun. 14, 3240 (2023).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  16. Villa, C. E. et al. CHD8 haploinsufficiency links autism to transient alterations in excitatory and inhibitory trajectories. Cell Rep. 39, 110615 (2022).

    Article  CAS  PubMed  Google Scholar 

  17. Warren, C. R. & Cowan, C. A. Humanity in a dish: population genetics with iPSCs. Trends Cell Biol. 28, 46–57 (2018).

    Article  CAS  PubMed  Google Scholar 

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

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  19. Kang, H. M. et al. Multiplexed droplet single-cell RNA-sequencing using natural genetic variation. Nat. Biotechnol. 36, 89–94 (2018).

    Article  CAS  PubMed  Google Scholar 

  20. Uzquiano, A. et al. Proper acquisition of cell class identity in organoids allows definition of fate specification programs of the human cerebral cortex. Cell 185, 3770–3788 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Dann, E., Henderson, N. C., Teichmann, S. A., Morgan, M. D. & Marioni, J. C. Differential abundance testing on single-cell data using k-nearest neighbor graphs. Nat. Biotechnol. 40, 245–253 (2022).

    Article  CAS  PubMed  Google Scholar 

  22. Cahill, K. M., Huo, Z., Tseng, G. C., Logan, R. W. & Seney, M. L. Improved identification of concordant and discordant gene expression signatures using an updated rank-rank hypergeometric overlap approach. Sci. Rep. 8, 9588 (2018).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  23. Trevino, A. E. et al. Chromatin and gene-regulatory dynamics of the developing human cerebral cortex at single-cell resolution. Cell 184, 5053–5069 (2021).

    Article  CAS  PubMed  Google Scholar 

  24. Polioudakis, D. et al. A single-cell transcriptomic atlas of human neocortical development during mid-gestation. Neuron 103, 785–801 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Cao, J. et al. The single-cell transcriptional landscape of mammalian organogenesis. Nature 566, 496–502 (2019).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  27. Manno, G. L. et al. RNA velocity of single cells. Nature 560, 494–498 (2018).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  28. Alfonso-Loeches, S. & Guerri, C. Molecular and behavioral aspects of the actions of alcohol on the adult and developing brain. Crit. Rev. Clin. Lab. Sci. 48, 19–47 (2011).

    Article  CAS  PubMed  Google Scholar 

  29. Arzua, T. et al. Modeling alcohol-induced neurotoxicity using human induced pluripotent stem cell-derived three-dimensional cerebral organoids. Transl. Psychiat. 10, 347 (2020).

    Article  CAS  Google Scholar 

  30. Carpita, B. et al. Autism spectrum disorder and fetal alcohol spectrum disorder: a literature review. Brain Sci. 12, 792 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Charness, M. E. Fetal alcohol spectrum disorders: awareness to insight in just 50 years. Alcohol Res. 42, 05 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  32. Eberhart, J. K. & Parnell, S. E. The genetics of fetal alcohol spectrum disorders. Alcohol Clin. Exp. Res. 40, 1154–1165 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Granato, A. & Dering, B. Alcohol and the developing brain: why neurons die and how survivors change. Int. J. Mol. Sci. 19, 2992 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  34. Marguet, F. et al. Oligodendrocyte lineage is severely affected in human alcohol-exposed foetuses. Acta Neuropathol. Commun. 10, 74 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Streissguth, A. P. & Dehaene, P. Fetal alcohol syndrome in twins of alcoholic mothers: concordance of diagnosis and IQ. Am. J. Med. Genet. 47, 857–861 (1993).

    Article  CAS  PubMed  Google Scholar 

  36. Sulik, K. K., Johnston, M. C. & Webb, M. A. Fetal alcohol syndrome: embryogenesis in a mouse model. Science 214, 936–938 (1981).

    Article  ADS  CAS  PubMed  Google Scholar 

  37. Meng, Q. et al. Human forebrain organoids reveal connections between valproic acid exposure and autism risk. Transl. Psychiatry 12, 130 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Church, G. M. The Personal Genome Project. Mol. Syst. Biol. 1, 2005.0030 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Sheridan, S. D. et al. Epigenetic characterization of the FMR1 gene and aberrant neurodevelopment in human induced pluripotent stem cell models of fragile X syndrome. PLoS ONE 6, e26203 (2011).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  40. Sugathan, A. et al. CHD8 regulates neurodevelopmental pathways associated with autism spectrum disorder in neural progenitors. Proc. Natl Acad. Sci. USA 111, E4468–E4477 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Thomson, J. A. et al. Embryonic stem cell lines derived from human blastocysts. Science 282, 1145–1147 (1998).

    Article  ADS  CAS  PubMed  Google Scholar 

  42. 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 

  43. Schindelin, J. et al. Fiji: an open-source platform for biological-image analysis. Nat. Methods 9, 676–682 (2012).

    Article  CAS  PubMed  Google Scholar 

  44. Goodchild, S. J. et al. Molecular pharmacology of selective NaV1.6 and dual NaV1.6/NaV1.2 channel inhibitors that suppress excitatory neuronal activity ex vivo. ACS Chem. Neurosci. 15, 1169–1184 (2024).

    Article  CAS  PubMed  Google Scholar 

  45. Zheng, G. X. Y. et al. Massively parallel digital transcriptional profiling of single cells. Nat. Commun. 8, 14049 (2017).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  46. Danecek, P. et al. Twelve years of SAMtools and BCFtools. GigaScience 10, giab008 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  47. Chen, Y., Lun, A. T. L. & Smyth, G. K. From reads to genes to pathways: differential expression analysis of RNA-seq experiments using Rsubread and the edgeR quasi-likelihood pipeline. F1000Research 5, 1438 (2016).

    PubMed  PubMed Central  Google Scholar 

  48. McCarthy, D. J., Chen, Y. & Smyth, G. K. Differential expression analysis of multifactor RNA-Seq experiments with respect to biological variation. Nucleic Acids Res. 40, 4288–4297 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Robinson, M. D., McCarthy, D. J. & Smyth, G. K. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 26, 139–140 (2010).

    Article  CAS  PubMed  Google Scholar 

  50. Cable, D. M. et al. Robust decomposition of cell type mixtures in spatial transcriptomics. Nat. Biotechnol. 40, 517–526 (2022).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank J. R. Brown for input and assistance in editing the manuscript; L. L. Lyons, N. Kozub and S. Tropp for technical assistance; all of the members of the Arlotta laboratory for discussions; B. Cohen for the Mito210 iPSC line; G. Church for the PGP1 iPSC line; M. Talkowski for the GM08330 iPSC line; the staff at the Broad Genomics Platform for sequencing; S. McCarroll for the Census-seq protocol; and B. Yeung for their support with pre-processing the Curioseeker data. Some components of the schematics were adapted from BioRender. This work was supported by grants from the Stanley Center for Psychiatric Research to P.A., R.N. and J.Z.L.; the Broad Institute of MIT and Harvard; the Blavatnik Biomedical Accelerator at Harvard University to P.A.; the National Institutes of Health (P50-MH094271 and RF1-MH123977 to P.A., R01-MH112940 to P.A. and J.Z.L., and U01-MH115727 to R.N.); and the Klarman Cell Observatory (A.R.).

Author information

Authors and Affiliations

Authors

Contributions

N.A.-B., I.F. and P.A. conceived and designed the experiments. N.A.-B., I.F. and S.A. generated, cultured and characterized all of the organoids in this study, with the help of S.T. and R.K., and P.A. supervised their work and contributed to data interpretation. N.A.-B., I.F., R.K. and S.A. performed IHC and image acquisition. R.K. performed MEA recordings and data analysis. X.A., N.A.-B. and I.F. performed scRNA-seq experiments, with help from D.J.D.B. and supervision of P.A. and J.Z.L.; N.A.-B., I.F. and S.T. performed scRNA-seq library preparation for the vast majority of the preparations included in this study. A.S.K., N.A.-B. and I.F. performed spatial transcriptomic experiments. A.S.K. and T.F. performed spatial transcriptomic computational analysis. A.W. performed the computational analysis of Extended Data Fig. 7. T.F., N.A.-B. and I.F. worked on cell type assignments and scRNA-seq data analysis, and T.F. performed all of the computational work under supervision by A.R. and P.A.; R.N. and M.T. provided the CW line, and advised on the PSC-Chimeroid experiments and on application of the Census-seq analysis pipeline, using computational tools developed by S. McCarroll’s laboratory. N.A.-B., I.F. and P.A. wrote the manuscript with contributions from all of the authors. All of the authors read and approved the final manuscript.

Corresponding author

Correspondence to Paola Arlotta.

Ethics declarations

Competing interests

P.A. is a scientific advisory board member at Foresite Labs, CNSII, and is a co-founder and scientific advisory board member of Vesalius Therapeutics. A.R. is a founder and equity holder of Celsius Therapeutics, an equity holder in Immunitas Therapeutics and, until 31 August 2020, was a scientific advisory board member of Syros Pharmaceuticals, Neogene Therapeutics, Asimov and Thermo Fisher Scientific. From 1 August 2020, A.R. has been an employee of Genentech and has equity in Roche.

Peer review

Peer review information

Nature thanks the anonymous reviewers 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.

Extended data figures and tables

Extended Data Fig. 1 PSC Chimeroids display uneven donor composition but proper cell type composition.

a, Schematic of the PSC-Chimeroid protocol. Some icons were created using BioRender. b, Stacked bar plots showing donor composition, as measured via Census-seq from low-pass whole-genome sequencing, in PSC-Chimeroids from various mixes and timepoints, labelled as the number of days since the day of the seeding (DIV0). For the DIV0 and DIV1 time points, n = 4 Chimeroids have been pulled together. c, Left panel: brightfield image of 3-mo PSC-Chimeroids; scale bar: 1 mm. Right panel: immunolabelling of 3-mo PSC-Chimeroid with SATB2, TBR1 and MAP2. Scale bar: 500 μm. d, UMAP of integrated PSC-Chimeroids at 3 mo, coloured by annotated cell type (left panel) and donor line as determined with demuxlet (right panel). e, UMAPs split by donor for two different replicates. f, Barplots of cell type proportions in PSC Chimeroids (n = 2), demultiplexed by donor. g, Immunolabelling of 1-mo PSC-Chimeroids showing early progenitors (SOX2), cortical progenitors (EMX1); rosette centres are lined with ZO-1, a tight junction protein present in the endfeet of radial glia cells, and NESTIN, indicating correctly-polarized neuroepithelium. CFuPNs neurons are marked by TBR1, and the neuronal dendrites by MAP2. h, Immunolabelling of 2-mo PSC-Chimeroids showing early progenitors (SOX2), IP (TBR2) and oRG (HOPX). CTNNB1 marks the centre of the neural rosettes, indicating correctly-polarized neuroepithelium. i, Immunolabelling of 3-mo PSC-Chimeroids showing cortical markers such as SATB2 (upper layers cortical neurons), and CTIP2 (deep layers cortical neurons). White arrows point to the neural rosettes (shown at higher magnification in the lower panels). Scale bar: 100 μm.

Extended Data Fig. 2 NSC-Chimeroids created by aggregating different numbers of cells display variable development.

a. Brightfield images of single-donor H1 NSC-Chimeroids seeded with 9,000 (upper) and 12,000 (lower) cells/well at day 3 and day 16 after reaggregation (left panels), and brightfield images of single donor PGP1 NSC-Chimeroids seeded with 9,000 (top left), 12,000 (bottom left), and 20,000 (top right) cells/well at day 3 and day 16 after reaggregation (right panels). Scale bar: 500 μm. Barplot showing growth over time of PGP1 NSC-Chimeroids compared across all different cell counts at aggregation (bottom right). Bars show median values, whiskers show upper and lower quartiles (n = 170 Chimeroids across all 4 timepoints). Significance was calculated using ANOVA followed by two-sided Tukey post-hoc pairwise tests. P-values: *** < − 0.001; **** - <0.0001. b. Immunolabelling of PGP1 single-donor NSC-Chimeroids made by aggregating the indicated number of cells, at DIV35. Upper images display whole organoids; lower images, enlargement of indicated portions. Lefthand panel, immunolabelling for SOX2 (progenitors), MAP2 (neuronal dendrites), and TBR1 (deep layer neurons). Righthand panel, immunolabelling for SOX2, ZO-1 (tight gap junctions), and EMX1 (dorsal cortical progenitors). Scale bar: 500 μm (upper) and 125 μm (lower). Arrowheads indicate non-cortical (grey) and cortical (white) regions. c. Left panel: whole-organoid brightfield images of H1 single-donor NPC-Chimeroids seeded with 100,000 cells at aggregation, at 18 and 35 days after mixing. Scale bar: 500 μm. Right panel: immunolabelling of H1 single-donor NPC-Chimeroids showing SOX2, ZO-1, and EMX1. Scale bar: 500 μm and 125 μm (zoom-in).

Extended Data Fig. 3 Multi-donor NSC Chimeroids display appropriate differentiation markers.

a, Immunolabelling of 1-mo MD-NSC Chimeroids showing early progenitors (SOX2), cortical progenitors (EMX1); rosette centres are lined with ZO-1, a tight junction protein present in the endfeet of radial glia cells, and NESTIN signal, indicating correctly-polarized neuroepithelium. CFuPNs neurons are marked by TBR1, and the neuronal dendrites by MAP2. b, Immunolabelling of 2-mo MD-NSC Chimeroids showing early progenitors (SOX2), IP (TBR2) and oRG (HOPX). CTNNB1 marks the centre of the neural rosettes, indicating correctly-polarized neuroepithelium. c, Immunolabelling of 3-mo MD-NSC Chimeroids showing cortical markers such as SATB2 (upper layers cortical neurons), and CTIP2 (deep layers cortical neurons). White arrows point to the neural rosettes. Scale bar: 100 μm. Some icons were created using BioRender.

Extended Data Fig. 4 NSC-Chimeroids develop appropriate cell-type composition across multiple donors.

a, Stacked barplots showing the donor composition, as measured via Census-seq from low-pass whole-genome sequencing, of multi-donor NSC-Chimeroids from various mixes and at various timepoints, labelled as the number of days since reaggregation (DIV; for the DIV0 and DIV1 time points, n = 4 chimeroids have been pulled together). b-e, 3-mo NSC-Chimeroids UMAPs split by donor (b, d) and stacked barplots of donor contributions for each cell type (c, e) for Mix 1 (4 donors) and Mix 2 (5 donors).

Extended Data Fig. 5 NSC-Chimeroids present uniform spatial distribution across donors.

a, Slice #1. Spatial plot of Slide-seq data from 2-mo MD-NSC Chimeroids (Mix 5), coloured by maximal donor contribution via Census-seq. b, Spatial plots showing donor prediction weights. c, Spatial plot of Slide-seq data from 2-mo NSC-Chimeroids (Mix 5), coloured by RCTD-assigned cell type. d, Spatial plots showing cell type prediction weights. e, Slice#2. Same as a. f, Slice#2. Same as b. g, Slide#2. Same as c. h, Slide#2. Same as d. i, Representative organoid placed on a Multielectrode Array (MEA). j, Representative activity map of a MD-NSC Chimeroid network burst at 4mo, where each pixel of the map represents an electrode (left) and example action potential traces from the electrodes highlighted in the activity map (right). k, Representative raster plot showing the firing activity of active electrodes (139, defined as electrodes with a mean firing rate >0.15 spikes/s inside the outline of the organoid) over a 15 min recording. Network bursts start to appear around 5 mins. l, Effect of AMPA and NMDA blockers (D-AP5, 150 µM; DNQX 60 µM) on neuronal activity/network bursts (left panel); network bursting activity of 4 recorded NSC-Chimeroids (right panel). Some icons were created using BioRender.

Extended Data Fig. 6 NPC-Chimeroids display appropriate differentiation markers.

a, Immunolabelling of 1-mo NPC-Chimeroids showing early progenitors (SOX2), cortical progenitors (EMX1); rosette centres are lined with ZO-1, a tight junction protein present in the endfeet of radial glia cells, and NESTIN signal, indicating correctly-polarized neuroepithelium. CFuPN neurons are marked by TBR1, and the neuronal dendrites by MAP2. b, Immunolabelling of 2-mo NPC-Chimeroids showing early progenitors (SOX2), IP (TBR2) and oRG (HOPX). CTNNB1 marks the centre of the neural rosettes, indicating correctly-polarized neuroepithelium. c, Immunolabelling of 3-mo NPC-Chimeroids showing cortical markers such as SATB2 (upper layers cortical neurons), and CTIP2 (deep layers cortical neurons). White arrows point to the neural rosettes. Scale bar: 100 μm. d, Brightfield images of 3-mo Chimeroids across all protocols (scale bar: 1 mm). Some icons were created using BioRender.

Extended Data Fig. 7 Nascent Chimeroid input to aggregation steps is composed of expected cell-type populations.

a, UMAP of integrated dataset containing DIV23 organoids from our reference map of Velasco-protocol organoid development (“Ref. Map”), and MD-NSC and MD-NPC-Chimeroids 1 day after aggregation (INPUT), colour-coded by annotated cell type. b. UMAPs split by protocol. c, UMAP of the INPUT for MD-NSC Chimeroids (mix 2), colour-coded by annotated cell type. d, UMAPs split by donors. e, UMAP of the INPUT for MD-NPC Chimeroids (Mix 6), colour-coded by annotated cell type. f, UMAPs split by donors. g, Donor demultiplexed after sc-RNA-seq. h, Bar plots of cell-type proportions in INPUT MD-NSC and MD-NPC Chimeroids for each donor. Some icons were created using BioRender.

Extended Data Fig. 8 Single-donor Chimeroids display appropriate differentiation markers.

a, Immunolabelling of 1-mo 11a SD-NSC Chimeroids showing early progenitors (SOX2), cortical progenitors (EMX1); rosette centres are lined with ZO-1, a tight junction protein present in the endfeet of radial glia cells, NESTIN and CTNNB1 signal, indicating correctly-polarized neuroepithelium surrounded by early new born CFuPN neurons. b, Same as panel a, for PGP1 SD-NSC Chimeroids. c, Immunolabelling of 3-mo 11a SD-NSC Chimeroids showing cortical markers such as SATB2 (upper layers cortical neurons), and CTIP2 (deep layers cortical neurons). d, Same as panel c, for PGP1 SD-NSC Chimeroids. White arrows point to the neural rosettes. Scale bar: 100 μm. Some icons were created using BioRender.

Extended Data Fig. 9 Chimeroids comparison across protocols and donors.

a, UMAP showing overlapping neighbourhoods of cells, as calculated using Milo. Red and blue colours indicate neighbourhoods with significant enrichment for cells from single-donor or multi-donor Chimeroids, respectively. Point size indicates the number of cells in a neighbourhood, and edge thickness indicates the number of cells shared between pairs of neighbourhoods. b, Beeswarm plot showing shifts in the composition of neighbourhoods of cells, grouped by the cell-type identity of those neighbourhoods. Each point represents a neighbourhood of 50-200 cells with similar gene expression profiles. The vertical axis indicates the enrichment of single-donor cells within a neighbourhood, with positive log fold change values indicating more than expected single-donor cells, and negative values indicating fewer than expected single-donor cells. Neighbourhoods are coloured based on statistical significance of that enrichment: grey, not significantly different from random; red, significant over-enrichment; blue, significant under-enrichment. c-d, Volcano plots showing DEGs, overall and for each donor (c), and for each major cell type (d), between the MD- and SD-NSC Chimeroid protocols. Positive log2 fold change indicates higher expression in the SD protocol. e, Correlation plots comparing the DEGs between each pair of donors in MD-NSC Chimeroids to those between the same pair of donors in the SD-NSC Chimeroids. The x- and y-axies shows log2 fold change in MD and SD, respectively. Point size and colour indicate statistical significance in MD and SD, respectively. f, Heatmap showing the mean absolute value of the log2 fold change between each donor in the MD-NSC Chimeroid protocol (columns) and each donor in the SD-NSC Chimeroid protocol (rows). Lower values imply more similarity; samples from each donor were transcriptionally most similar to samples from the same donor in the other protocol. g, Aitchison distance measuring the dissimilarity in cell type composition between replicates within each protocol, split into comparisons within batches and between batches, and limited to cells derived from the PGP1 and Mito210 donors (as only these two donors are present in all organoid/Chimeroid protocols assayed here). (n = 23 PGP1, 23 Mito210, and 10 fetal samples; Boxes show upper and lower quartiles and median, whiskers show highest/lowest values within 1.5 interquartile range (IQR) of the nearest hinge). The dissimilarity in cell type composition between samples within each of three fetal cortical datasets is also shown, indicating natural variability between individuals. Dotted lines represent mean inter-sample distances within each fetal dataset20,23,24. Some icons were created using BioRender.

Extended Data Fig. 10 Chimeroids display similar cell type complexity as single-donor organoids and fetal tissue.

Rank-Rank Hypergeometric Overlap (RR-HO) plots comparing the expression signatures of cell types in multi-donor Chimeroids to all cell types in single-donor Chimeroids, cortical organoids from our reference map of Velasco-protocol organoid development, or endogenous human fetal tissue20. The horizontal axes represent lists of marker genes for multi-donor NSC-Chimeroid cell types compared to all other Chimeroid cells, ranked from most upregulated to most downregulated; the vertical axes represent similarly-ranked lists of marker genes for single-donor NSC-Chimeroids, reference map organoids, or human fetal cells. Colour at a given position represents the significance (negative log p-value) of the overlap of the gene lists up to that point, as calculated by Fisher’s exact tests. High significance (i.e., red colour) in the lower left and upper right quadrants indicates strong concordance between the expression profiles which define the compared cell types. a, b, c, d and e represent different cell types analysed. f, Explanatory schematic for the RR-HO plots. Some icons were created using BioRender.

Extended Data Fig. 11 Chimeroids display similar developmental trajectories and metabolic hallmarks across donors and across protocols.

a, Density plot showing the distribution of scaled pseudotimes assigned to each donor within each organoid/Chimeroid protocol. Pseudotime was calculated independently for each protocol using Monocle 3, with aRGs in each dataset set as root cells. b, Glycolysis module scores calculated in each cell type/donor/Chimeroid with linear MEMs (using lme4’s lmer) on the PGP1/Mito210 cells using donor as a random effect (n = 40 replicates across 4 protocols; boxes show upper and lower quartiles and median, whiskers show highest/lowest values within 1.5 interquartile range (IQR) of the nearest hinge.). P-values were generated via one-sided F-test comparing models with and without “protocol” as a covariate. c-d, Expression of the glycolysis geneset is similar across donors (12,887 cells from n = 45 donor/Chimeroids) and protocols (20,425 cells from n = 40 donor/Chimeroids and organoids). Violin plots showing the module scores for the MSigDB Hallmark Glycolysis gene set across cell types, donors, and protocols; boxes show upper and lower quartiles and median, whiskers show highest/lowest values within 1.5 interquartile range (IQR) of the nearest hinge. Module scores were calculated with Seurat’s addModuleScores function.

Extended Data Fig. 12 VPA and EtOH treatment differently affect cell-type composition in NSC-Chimeroids.

a, Stacked barplot showing cell type and donor composition for control Chimeroids; the width of each bar corresponds to the proportion of the indicated donor in Mix 1 (4 donors, left panel) and Mix 2 (5 donors, right panel). b, Brightfield images of whole cortical NSC-Chimeroids at 3 mo, in the EtOH treatment condition. Scale bar: 1 mm. c, Stacked barplot showing cell type and donor composition for EtOH treated Chimeroids; the width of each bar corresponds to the proportion of the indicated donor in Mix 1 (4 donors, left panel) and Mix 2 (5 donors, right panel). d, UMAPs of EtOH treated NSC-Chimeroids, split by mixes and replicates. e, Brightfield images of whole cortical MD-NSC Chimeroids at 3 mo, in the VPA treatment condition. Scale bar: 1 mm. f, Stacked barplot showing cell type and donor composition for EtOH treated Chimeroids; the width of each bar corresponds to the proportion of the indicated donor in Mix 1 (4 donors, upper panel) and Mix 2 (5 donors, lower panel). g, UMAPs of VPA treated NSC-Chimeroids, split by mixes and replicates. Some icons were created using BioRender.

Extended Data Fig. 13 VPA treatment cause protein and transcriptome alterations in single-donor Chimeroids.

a, Immunolabelling of multi donor NSC-Chimeroids (a, Mix 1, upper images and b, Mix 2, lower images). Left panel, MAP2 (neuronal dendrites), EMX1 (cortical progenitors), and DLX2 (GABAergic cells). Right panel, DCX (migrating neurons), SATB2 (upper layers cortical neurons), and CTIP2 (deep layers cortical neurons). Scale bar: 100 μm. c, Stacked barplot showing cell type composition for control and treated single-donor NSC-Chimeroids. d, Left panel: cell-type specific changes in single-donor Chimeroids treated with VPA (yellow) vs control (grey). Right panel: UMAP showing overlapping neighbourhoods of cells, as calculated using Milo. Red and blue colours indicate neighbourhoods with significant enrichment for VPA-treated cells or control cells, respectively. Point size indicates the number of cells in a neighbourhood, and edge thickness indicates the number of cells shared between pairs of neighbourhoods. e, Beeswarm plot showing shifts in the composition of neighbourhoods of cells in response to VPA treatment in single-donor Chimeroids, grouped by the cell-type identity of those neighbourhoods. Each point represents a neighbourhood of 50-200 cells with similar gene expression profiles. The vertical axis indicates the enrichment of VPA-treated cells within a neighbourhood, with positive log fold change values indicating more than expected VPA-treated cells, and negative values indicating fewer than expected VPA-treated cells. Neighbourhoods are coloured based on statistical significance of that enrichment: grey, not significantly different from random; red, significant over-enrichment; blue, significant under-enrichment. If most neighbourhoods within a cell type collectively shift up or down, it implies an overall gain or loss, respectively, of that cell type in VPA-treated Chimeroids. Cell types with neighbourhoods that form long tails of both over-enrichment and under-enrichment are likely to have treatment-induced changes in expression profile, without necessarily changing in abundance. f, GSEA of donor specific genes from MD-NSC Chimeroids ranked on the corresponding single-donor datasets. g, GSEA of donor specific genes from SD-NSC Chimeroids ranked on the corresponding multi-donor datasets (lower panels). P-values calculated via two-sided Kolmogrov-Smirnov test. Some icons were created using BioRender.

Supplementary information

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

Antón-Bolaños, N., Faravelli, I., Faits, T. et al. Brain Chimeroids reveal individual susceptibility to neurotoxic triggers. Nature 631, 142–149 (2024). https://doi.org/10.1038/s41586-024-07578-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-024-07578-8

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