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:

An in vivo model of functional and vascularized human brain organoids

An Erratum to this article was published on 06 August 2018

This article has been updated

Abstract

Differentiation of human pluripotent stem cells to small brain-like structures known as brain organoids offers an unprecedented opportunity to model human brain development and disease. To provide a vascularized and functional in vivo model of brain organoids, we established a method for transplanting human brain organoids into the adult mouse brain. Organoid grafts showed progressive neuronal differentiation and maturation, gliogenesis, integration of microglia, and growth of axons to multiple regions of the host brain. In vivo two-photon imaging demonstrated functional neuronal networks and blood vessels in the grafts. Finally, in vivo extracellular recording combined with optogenetics revealed intragraft neuronal activity and suggested graft-to-host functional synaptic connectivity. This combination of human neural organoids and an in vivo physiological environment in the animal brain may facilitate disease modeling under physiological conditions.

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

Figure 1: Intracerebral grafting of brain organoids into mouse brain.
Figure 2: Survival and differentiation of grafted organoids.
Figure 3: Organoid graft sends axonal projections with synaptic connectivity.
Figure 4: Functional vasculature and decreased apoptosis in grafts.
Figure 5: Two-photon imaging reveals neuronal activity in the graft.
Figure 6: In vivo electrophysiological recording in the graft.

Similar content being viewed by others

Change history

  • 18 July 2018

    In the version of this article initially published, credit for part of Figure 4a was omitted. The image of a lens and microscope stage was originally published elsewhere. The error has been corrected in the HTML and PDF versions of the article.

References

  1. Mertens, J., Marchetto, M.C., Bardy, C. & Gage, F.H. Evaluating cell reprogramming, differentiation and conversion technologies in neuroscience. Nat. Rev. Neurosci. 17, 424–437 (2016).

    Article  CAS  Google Scholar 

  2. Shanks, N., Greek, R. & Greek, J. Are animal models predictive for humans? Philos. Ethics Humanit. Med. 4, 2 (2009).

    Article  Google Scholar 

  3. Kelava, I. & Lancaster, M.A. Dishing out mini-brains: current progress and future prospects in brain organoid research. Dev. Biol. 420, 199–209 (2016).

    Article  CAS  Google Scholar 

  4. 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  Google Scholar 

  5. Sasai, Y., Eiraku, M. & Suga, H. In vitro organogenesis in three dimensions: self-organising stem cells. Development 139, 4111–4121 (2012).

    Article  CAS  Google Scholar 

  6. Mariani, J. et al. Modeling human cortical development in vitro using induced pluripotent stem cells. Proc. Natl. Acad. Sci. USA 109, 12770–12775 (2012).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  9. 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 

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

    Article  CAS  Google Scholar 

  11. Jo, J. et al. Midbrain-like organoids from human pluripotent stem cells contain functional dopaminergic and neuromelanin-producing neurons. Cell Stem Cell 19, 248–257 (2016).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  14. 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 

  15. Bershteyn, M. et al. Human iPSC-derived cerebral organoids model cellular features of lissencephaly and reveal prolonged mitosis of outer radial glia. Cell Stem Cell 20, 435–449.e4 (2017).

    Article  CAS  Google Scholar 

  16. Iefremova, V. et al. An organoid-based model of cortical development identifies non-cell-autonomous defects in Wnt signaling contributing to Miller-Dieker syndrome. Cell Rep. 19, 50–59 (2017).

    Article  CAS  Google Scholar 

  17. Shen, Q. et al. Endothelial cells stimulate self-renewal and expand neurogenesis of neural stem cells. Science 304, 1338–1340 (2004).

    Article  CAS  Google Scholar 

  18. Yin, X. et al. Engineering stem cell organoids. Cell Stem Cell 18, 25–38 (2016).

    Article  CAS  Google Scholar 

  19. Giandomenico, S.L. & Lancaster, M.A. Probing human brain evolution and development in organoids. Curr. Opin. Cell Biol. 44, 36–43 (2017).

    Article  CAS  Google Scholar 

  20. Schwartz, M.P. et al. Human pluripotent stem cell-derived neural constructs for predicting neural toxicity. Proc. Natl. Acad. Sci. USA 112, 12516–12521 (2015).

    Article  CAS  Google Scholar 

  21. Stenevi, U., Björklund, A. & Svendgaard, N.A. Transplantation of central and peripheral monoamine neurons to the adult rat brain: techniques and conditions for survival. Brain Res. 114, 1–20 (1976).

    Article  CAS  Google Scholar 

  22. Gage, F.H. & Björklund, A. Intracerebral grafting of neuronal cell suspensions into the adult brain. Cent. Nerv. Syst. Trauma 1, 47–56 (1984).

    Article  CAS  Google Scholar 

  23. Yui, S. et al. Functional engraftment of colon epithelium expanded in vitro from a single adult Lgr5+ stem cell. Nat. Med. 18, 618–623 (2012).

    Article  CAS  Google Scholar 

  24. Watson, C.L. et al. An in vivo model of human small intestine using pluripotent stem cells. Nat. Med. 20, 1310–1314 (2014).

    Article  CAS  Google Scholar 

  25. Takebe, T. et al. Vascularized and functional human liver from an iPSC-derived organ bud transplant. Nature 499, 481–484 (2013).

    Article  CAS  Google Scholar 

  26. Clevers, H. Modeling development and disease with organoids. Cell 165, 1586–1597 (2016).

    Article  CAS  Google Scholar 

  27. Kretzschmar, K. & Clevers, H. Organoids: modeling development and the stem cell niche in a dish. Dev. Cell 38, 590–600 (2016).

    Article  CAS  Google Scholar 

  28. O'Rourke, K.P. et al. Transplantation of engineered organoids enables rapid generation of metastatic mouse models of colorectal cancer. Nat. Biotechnol. 35, 577–582 (2017).

    Article  CAS  Google Scholar 

  29. Roper, J. et al. In vivo genome editing and organoid transplantation models of colorectal cancer and metastasis. Nat. Biotechnol. 35, 569–576 (2017).

    Article  CAS  Google Scholar 

  30. Dye, B.R. et al. A bioengineered niche promotes in vivo engraftment and maturation of pluripotent stem cell derived human lung organoids. eLife 5, e19732 (2016).

    Article  Google Scholar 

  31. Gage, F.H. & Fisher, L.J. Intracerebral grafting: a tool for the neurobiologist. Neuron 6, 1–12 (1991).

    Article  CAS  Google Scholar 

  32. Gage, F.H., Björklund, A. & Stenevi, U. Denervation releases a neuronal survival factor in adult rat hippocampus. Nature 308, 637–639 (1984).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  34. 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  Google Scholar 

  35. 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  Google Scholar 

  36. Dana, H. et al. Sensitive red protein calcium indicators for imaging neural activity. eLife 5, e12727 (2016).

    Article  Google Scholar 

  37. Jin, X. & Costa, R.M. Start/stop signals emerge in nigrostriatal circuits during sequence learning. Nature 466, 457–462 (2010).

    Article  CAS  Google Scholar 

  38. Falkner, S. et al. Transplanted embryonic neurons integrate into adult neocortical circuits. Nature 539, 248–253 (2016).

    Article  Google Scholar 

  39. Golshani, P. et al. Internally mediated developmental desynchronization of neocortical network activity. J. Neurosci. 29, 10890–10899 (2009).

    Article  CAS  Google Scholar 

  40. Espuny-Camacho, I. et al. Pyramidal neurons derived from human pluripotent stem cells integrate efficiently into mouse brain circuits in vivo. Neuron 77, 440–456 (2013).

    Article  CAS  Google Scholar 

  41. Grealish, S. et al. Monosynaptic tracing using modified rabies virus reveals early and extensive circuit integration of human embryonic stem cell-derived neurons. Stem Cell Rep. 4, 975–983 (2015).

    Article  CAS  Google Scholar 

  42. Cunningham, M. et al. hPSC-derived maturing GABAergic interneurons ameliorate seizures and abnormal behavior in epileptic mice. Cell Stem Cell 15, 559–573 (2014).

    Article  CAS  Google Scholar 

  43. Qi, Y. et al. Combined small-molecule inhibition accelerates the derivation of functional cortical neurons from human pluripotent stem cells. Nat. Biotechnol. 35, 154–163 (2017).

    Article  CAS  Google Scholar 

  44. Thompson, L.H. & Björklund, A. Reconstruction of brain circuitry by neural transplants generated from pluripotent stem cells. Neurobiol. Dis. 79, 28–40 (2015).

    Article  CAS  Google Scholar 

  45. Kriks, S. et al. Dopamine neurons derived from human ES cells efficiently engraft in animal models of Parkinson's disease. Nature 480, 547–551 (2011).

    Article  CAS  Google Scholar 

  46. Bachoud-Lévi, A.C. & Perrier, A.L. Regenerative medicine in Huntington's disease: current status on fetal grafts and prospects for the use of pluripotent stem cell. Rev. Neurol. (Paris) 170, 749–762 (2014).

    Article  Google Scholar 

  47. Barker, R.A., Drouin-Ouellet, J. & Parmar, M. Cell-based therapies for Parkinson disease—past insights and future potential. Nat. Rev. Neurol. 11, 492–503 (2015).

    Article  CAS  Google Scholar 

  48. Lindvall, O. Clinical translation of stem cell transplantation in Parkinson's disease. J. Intern. Med. 279, 30–40 (2016).

    Article  CAS  Google Scholar 

  49. Steinbeck, J.A. & Studer, L. Moving stem cells to the clinic: potential and limitations for brain repair. Neuron 86, 187–206 (2015).

    Article  CAS  Google Scholar 

  50. Kikuchi, T. et al. Human iPS cell-derived dopaminergic neurons function in a primate Parkinson's disease model. Nature 548, 592–596 (2017).

    Article  CAS  Google Scholar 

  51. 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  CAS  Google Scholar 

  52. Perrin, S. Preclinical research: make mouse studies work. Nature 507, 423–425 (2014).

    Article  Google Scholar 

  53. Mak, I.W., Evaniew, N. & Ghert, M. Lost in translation: animal models and clinical trials in cancer treatment. Am. J. Transl. Res. 6, 114–118 (2014).

    PubMed  PubMed Central  Google Scholar 

  54. Assawachananont, J. et al. Transplantation of embryonic and induced pluripotent stem cell-derived 3D retinal sheets into retinal degenerative mice. Stem Cell Rep. 2, 662–674 (2014).

    Article  Google Scholar 

  55. Huch, M. et al. Unlimited in vitro expansion of adult bi-potent pancreas progenitors through the Lgr5/R-spondin axis. EMBO J. 32, 2708–2721 (2013).

    Article  CAS  Google Scholar 

  56. Huch, M., Boj, S.F. & Clevers, H. Lgr5(+) liver stem cells, hepatic organoids and regenerative medicine. Regen. Med. 8, 385–387 (2013).

    Article  CAS  Google Scholar 

  57. Huch, M. et al. In vitro expansion of single Lgr5+ liver stem cells induced by Wnt-driven regeneration. Nature 494, 247–250 (2013).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  59. Mansour, A.A., Khazanov-Zisman, S., Netser, Y., Klar, A. & Ben-Arie, N. Nato3 plays an integral role in dorsoventral patterning of the spinal cord by segregating floor plate/p3 fates via Nkx2.2 suppression and Foxa2 maintenance. Development 141, 574–584 (2014).

    Article  CAS  Google Scholar 

  60. Gonçalves, J.T., Anstey, J.E., Golshani, P. & Portera-Cailliau, C. Circuit level defects in the developing neocortex of Fragile X mice. Nat. Neurosci. 16, 903–909 (2013).

    Article  Google Scholar 

  61. Gonçalves, J.T. et al. In vivo imaging of dendritic pruning in dentate granule cells. Nat. Neurosci. 19, 788–791 (2016).

    Article  Google Scholar 

  62. Heiney, S.A., Wohl, M.P., Chettih, S.N., Ruffolo, L.I. & Medina, J.F. Cerebellar-dependent expression of motor learning during eyeblink conditioning in head-fixed mice. J. Neurosci. 34, 14845–14853 (2014).

    Article  CAS  Google Scholar 

  63. Pologruto, T.A., Sabatini, B.L. & Svoboda, K. ScanImage: flexible software for operating laser scanning microscopes. Biomed. Eng. Online 2, 13 (2003).

    Article  Google Scholar 

  64. Jin, X., Tecuapetla, F. & Costa, R.M. Basal ganglia subcircuits distinctively encode the parsing and concatenation of action sequences. Nat. Neurosci. 17, 423–430 (2014).

    Article  CAS  Google Scholar 

  65. Perkel, D.H., Gerstein, G.L. & Moore, G.P. Neuronal spike trains and stochastic point processes. II. Simultaneous spike trains. Biophys. J. 7, 419–440 (1967).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank members of the Gage laboratory for helpful discussions; S. Schafer for the pCSC-CAG-GFP lentivirus and I. Verma for the pBOB-CAG-Td-Tomato construct. We also thank M. Shtrahman for assistance and two-photon imaging expertise, L. Moore, S. Baktvar, S. Kim, B. Miller, C. Lim, and I. Guimont for their technical assistance, M.L. Gage for editorial comments, V. Mertens for illustrations, I. Farhy-Tselnicker and J. Klug for technical advice, and M. Shtrahman, and T. Toda for critical reading and comments on the manuscripts. We thank U. Manor and the Waitt Advanced Biophotonics Core, K. Diffenderfer and the Salk Stem Cell Core, C. O'Connor and C. Fitzpatrick and the FACS Core, and the Salk Institute for generously providing critical infrastructural and financial support. We apologize to those whose work was not cited owing to space limitations. This work was supported by the NIH (U19 MH106434, U01 MH106882), The Paul G, Allen Family Foundation, Bob and Mary Jane Engman, The Leona M, and Harry B, Helmsley Charitable Trust Grant (2012-PG-MED), Annette C, Merle-Smith, The G, Harold and Leila Y, Mathers Foundation, JPB Foundation, Dolby Family Ventures for F.H.G. and NIH grants (R01NS083815, R01AG047669) for X.J. S.F. was funded by CIRM Bridges to Stem Cell Research Internship Program. A.A.M. received funding from the EMBO Postdoctoral Long-term Fellowship (ALTF 1214-2014, EMBO fellowship is co-funded also by the European Commission FP7-Marie Curie Actions, LTFCOFUND2013, GA-2013-609409), and is currently supported by the Human Frontiers Science Program (HFSP Long-Term Fellowship- LT001074/2015).

Author information

Authors and Affiliations

Authors

Contributions

A.A.M. and F.H.G. conceived the idea for the project and wrote the manuscript. A.A.M. designed and performed the experiments and analyzed the data. A.A.M. generated hESC lines and organoids culture, performed cellular, molecular and histological assays and analyzed the data. S.F. and D.Q. performed cell culture and histological experiments under the supervision of A.A.M. A.A.M., C.W.B., F.H.G., J.T.G., and S.J. performed surgeries. T.G. and C.W.B. performed two-photon imaging and analyzed the data. S.L.P. performed behavioral experiments. H.L. performed electrophysiological microelectrode and optogenetic experiments under the supervision of X.J. F.H.G. supervised the project and provided funding.

Corresponding author

Correspondence to Fred H Gage.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Generation and characterization of GFP+ cerebral organoids.

(a) Microscopic image shows a single colony of H9-GFP+ hESCs (Left), and a representative example of 46-days old GFP+ brain organoids used for the transplantation experiments. (b) Immunofluorescence staining for GFP of GFP-expressing cerebral organoids at day 60. Note, unlike the perimeter areas, the center of the organoid­­ lacks GFP expression. Solid line indicates the border of the low-GFP expressing region. (c) Cerebral organoids immunostained for PAX6 and CTIP2 at 38 days. (d) 38-days old cerebral organoids immunostained for SOX2 and NeuN. Right panel is magnification of the boxed area in the left panel. Scale bar is 1 mm in a, 200 μm in b, 20 μm in c, d (right), and 200 μm in d (left).

Supplementary Figure 2 In vivo long-term survival of grafted brain organoids.

(a) Immunofluorescence staining for GFP and human mitochondria (hMito), which specifically labels human cells, of the grafted brain organoid at 14 dpi. Insets show staining form the same section of neurites projection into the host brain. (b) Lower power slide scanner image of a coronal brain section stained with anti-GFP antibody showing robust integration of GFP+ human brain organoids at 90 dpi. Unlike organoids grown in culture before grafting (Supplementary Fig. 1b), there are no detectable signs of regions that lack GFP expression in the center of the organoid 3 months post-implantation. (c) Left, confocal stitched tile scan of 90dpi-organoid graft stained with GFP. Right, Histogram of GFP intensity at various positions of the graft across the red line. Nuclei were counterstained with DAPI. Scale bar is, 100 μm in a, 1 mm in b, 200 μm in c.

Supplementary Figure 3 In vivo neuronal differentiation of grafted brain organoids.

(a) Immunofluorescence staining for the mature astrocyte marker GFAP (left) and microglial marker Iba1 (right) on a whole organoid cross section at 46 days in culture. (b) Immunofluorescence staining of the mature neuronal dendrite marker MAP2 (left panel), astrocyte marker S100B (middle panel), and triple staining of GFP, hGFAP and SOX2 (right panel) inside the organoid graft at 50 dpi. Note the cellular colocalization between hGFAP and SOX2; co-association of SOX2 and hGFAP suggests that SOX2+ cells are glia and not NPCs.

(c) Immunofluorescence staining of GFP, Sox2, and NeuN within the grafted brain organoid at 90 dpi.(d) Immunofluorescence staining of GFP and MAP2 within the grafted brain organoid at 90 dpi. (e) Immunofluorescence staining of GFP, hNuclei, and SOX2 within the grafted brain organoid at 233 dpi. (f) Immunofluorescence staining of GFP and NeuN, within the grafted organoid at 233 dpi. (g) Immunofluorescence staining of GFP, Olig2, and Iba1 within the grafted brain organoid at 233 dpi. (h) Example image of coronal section obtained from grafted mouse brain at 90 dpi. Nuclei were counterstained with DAPI. Scale bar is, 500 μm in a, 50 μm in b-g, 1 mm in h.

Supplementary Figure 4 Brain organoids send long-distance axonal projections with synaptic connectivity after implantation in mouse brain.

(a) Slide scanner images of coronal brain sections stained for GFP and obtained from 90-dpi grafted mouse brain show robust integration of GFP+ organoids and very large numbers of axons extending into the grafting region and into more rostral section of the host brain. Dashed boxes indicate the sampled region from which higher magnification views in (b) were obtained. Note that images were over-saturated for the signal in the graft region to show the GFP signal in the axons trajectories. Note that the high background on the edge of the section is a nonspecific signal. Scale bars:1 mm in a and 200 μm in b.

Supplementary Figure 5 Vascularization and two-photon imaging of the organoid graft.

(a) Double immunofluorescence staining of GFP and the endothelial marker Endoglin in the indicated post-implantation stage showing the intensive growth of blood vessels inside the organoid graft. (b) Double immunofluorescence staining for human-specific CD31 and CD31 (recognizing both mouse and human) of organoid graft harvested at 90 dpi. Note that the organoid graft is negative for human CD31, suggesting a host origin of the infiltrated blood vessels at the examined time point. (c) Quantification of vascularization success rate in the grafted organoids. Average value is 85.4%±6.4; data represent mean±s.e.m (n=10 independent experiments, total of 55 animals). (d) Surface area change of single grafted organoids during the first 2 weeks of implantation. (e) The average change of total organoid surface area during the first 2 weeks of implantation normalized to day 0 (100%). Data represent mean ± s.e.m from at least 3 grafted animals; day 0-5 (n=9), day 6 (n=8), day 7 and 8 (n=6), day 13 (n=4), day 14 (n=3). (f) Deep in vivo two-photon imaging of GFP signal of the implanted organoids in a head-fixed awake mouse through the cranial window, demonstrating the feasibility of the imaging system. Image shows maximum projection of 300 μm from 200-500 μm inside the grafted organoid from a 30 dpi mouse. (g) Two-photon imaging of blood vessels inside the grafted organoid. Dextran was infused in engrafted animal at 120 dpi. Single z-plane acquired at 418 μm depth below the organoid surface, showing blood flow inside the vascular network (see Supplementary video 4). Scale bar is 50 μm in a,b and 100 μm in h,g.

Supplementary Figure 6 Electrophysiological recording of neuronal activity in the grafted brain organoids.

(a) Firing rate changes of single neurons obtained from 50-dpi organoid at two different levels of depth (top: −0.9 mm; bottom: −1.0 mm) from the graft surface. Each line (color-coded) indicates the firing rate of an individual neuron. Arrows on the top denote the time isoflurane was turned ON (filled arrows) and OFF (empty arrows). (b) Spike raster plots from the neurons in (a) at two different depths (top: −0.9 mm; bottom: −1.0 mm). Each vertical bar indicates a single spike. (c) Cross-correlation of neuron pairs in (a) at depths of −0.9 mm (top) and −1.0 mm (bottom). (d,e) Example of cross correlation analysis performed on one neuron pair from a 115 dpi organoid graft (Fig. 6f). Analysis was performed by testing the significance of the central peak observed in cross-correlation (d). The threshold for the test was calculated based on mean + 3*SD of the baseline, which was defined as the time window from −100 ms to −20 ms in cross-correlation (d, dashed blue line). Obviously, although the magnitude is relatively small, the central peak significantly exceeds the threshold, which is evident of correlation between neurons. Note that when shuffling the neuronal activity with a random duration, the shuffled cross-correlation between the same neurons in (d) became completely flat with no peak (e), indicating that the correlation is indeed from the correlated activity, rather than noise. (f) Spikes with magnitude exceeding the threshold were tagged and recorded as waveforms (red arrows). Then we applied PCA analysis to these waveforms to discriminate the neuronal spikes from noise. On PC plane, waveforms showing distinct shapes are classified into separate clusters.

Supplementary Figure 7 Electrophysiological recording of neuronal activity in the grafted brain organoids.

Examples of in vivo recording from the organoid graft and the host brain. (a-b) Left panels, firing rate changes of single neurons obtained from the indicated time points and the dorsal-ventral (DV) depth from the graft surface. Each line (color-coded) indicates the firing rate of an individual neuron. Arrows on the top denote the time isoflurane was turned ON (filled arrows) and OFF (empty arrows). Right panels, spike raster plots from the neurons in the left panel. Each vertical bar indicates a single spike. (c) Firing rate changes of single neurons obtained from the host cortical region.

Supplementary Figure 8 Transduction of ChR2 into the grafted organoid for optogenetic stimulation.

Organoid graft was injected with AAV-hSyn-ChR2-YFP and analyzed at 155 dpi. (a) Immunostaining for GFP (labeling hSyn::ChR2-YFP) and hNuclei inside the organoid graft. (b) Immunostaining for tdTomato (labeling organoid graft) and GFP (labeling hSyn::ChR2-YFP) in the cortex of the host brain at 155 dpi. Bottom panels are magnification of the boxed area in the top panel. Note the fragmented tdTomato expression along the axons perhaps due to long term expression. Nuclei were counterstained with DAPI. Scale bar is 50 μm.

Supplementary Figure 9 ptogenetic stimulation evokes neuronal responses in the organoid-graft expressing ChR2.

(a) Diagram of optogenetic stimulation and electrophysiological recording in un-injected control organoid-graft. (b) Light-evoked responses from the same single neuron to 1s constant (left panel), 50 Hz (middle panel), or 20 Hz (right panel) light stimulation in control graft. Top panels: raster plot of neuronal responses in 30 trials. Each tick indicates a single spike. Bottom panels: PETH of the average firing rate across 30 trials. (c,d) Organoid graft was injected with AAV-hSyn-ChR2-YFP and analyzed at 155dpi. (c) Diagram of optogenetic stimulation and electrophysiological recording in organoid graft. (d) Light-evoked responses from the same single neuron to 1s constant (left panel), 50 Hz (middle panel), or 20 Hz (right panel) light stimulation. The stimulation onset is at 0. Top panels: raster plot of neuronal responses in 30 trials. Each tick indicates a single spike. Bottom panels: PETH of the average firing rate across 30 trials. The response latency to the laser onset was less than 5 milliseconds and no detectable response was observed before or after the period of light illumination. n= 2 animals. All recordings were performed while animals were under isoflurane anesthesia.

Supplementary Figure 10 Optogenetic stimulation reveals intragraft neuronal activity and functional output connectivity from the organoid graft to host brain.

(a) Diagram of optogenetic stimulation in tdTomato-expressing uninfected-control organoid and electrophysiological recording in the host brain. (b) Local field potential (LFP) recorded from a single electrode in the control host brain after optogenetic stimulation of organoid with 20 Hz light stimulation. The voltage is color coded for 30 trials (top). Averaged LFP across trials (bottom). (c) LFP changes recorded from a different electrode from the same brain region in (b). (d-h) Organoid graft was injected with AAV-hSyn-ChR2-YFP and analyzed at 155dpi. (d) Diagram of optogenetic stimulation in tdTomato-expressing, AAV-hSyn-ChR2-YFP-infected, organoid and electrophysiological recording in the host brain (fiber location: DV -1.7mm; array location: AP -2.54 mm, ML -1.5 mm, DV -2.2 mm). (e) Local field potential recorded from a single electrode in the host brain region. Optogenetic stimulation of organoid with 20 Hz drives LFP changes in the host brain region. The voltage is color coded for 30 trials (top). Averaged LFP across trials (bottom). Inset: Averaged LFP with a finer time scale. (f) Power spectral density of the averaged LFP in (e). (g) LFP changes recorded from a different electrode from the same brain region in (e). Inset: Averaged LFP with a finer time scale. (h) Power spectral density of the averaged LFP in (g). (i-k) LFP changes recorded from the same animal and brain region as in (d) but from different, more dorsal array location (AP -2.54 mm, ML -1.5 mm, DV -0.8mm). n=2 animals. All recordings were performed while animals were under isoflurane anesthesia.

Supplementary Figure 11 Behavioral examination of spatial learning abilities in organoid-grafted mice

(a) Latency to locate target during Barnes maze training of 3 trials per day for 3 days. Values for individual mice were averaged within each day for analysis. Training performance was analyzed by two-way ANOVA with group as a between subject factor and day as a within-subject factor. There was a significant effect of day (F(2,24) = 24.3, p < 0.0001), but no effect of group (F(1,12) = 0.8116, p = 0.3854) or group X day interaction (F(2,24) = 2.878, p = 0.0758). (b) Latency to locate target during Barnes maze probe trial on day 4, 24 hr after final training day. Probe trial performance was analyzed by the Mann-Whitney test. Unengrafted vs. grafted p = 0.1474. (c) Errors prior to locating target during Barnes maze training of 3 trials per day for 3 days. Values for individual mice were averaged within each day for analysis. There was a significant effect of day (F(2,24) = 7.244, p = 0.0035) and a group X day interaction (F(2,24) = 7.878, p = 0.0023; **post-hoc Sidak’s multiple comparison test p = 0.0052 for training Day 1, p = 0.7258 for Day 2, p = 0.9438 for Day 3). No main effect was observed for group (F(1,12) = 0.8008, p = 0.3884). (d) Pokes to target location during Barnes maze probe trial on day 4, 24 hr after final training day. **Unengrafted vs. grafted p = 0.0076 (e) Location of errors during probe trial for Nod-Scid mice. (f) Location of errors during probe trial for organoid implanted mice. Values in a-f represent mean ± s.e.m. for each group, n=7 mice per group.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–11 (PDF 2391 kb)

Life Sciences Reporting Summary (PDF 162 kb)

Supplementary Table 1

List of primary and secondary antibodies. (PDF 81 kb)

Two-photon in vivo imaging of organoid graft in awake, head-fixed mouse.

GFP+ organoid (green) was grafted into mouse brain and was imaged under a two-photon microscope at 59 dpi. Video shows serial images acquired from 1 to 500 μm below the organoid surface. Note that higher laser power (compared to other experiments) was used to achieve significant signal upon penetration to 500 μm. (AVI 40870 kb)

Two-photon in vivo imaging of blood vessels inside the organoids.

GFP+ organoid (green) was grafted into mouse brain and the mouse was injected with Dextran (red) at 30 dpi, and imaged directly. Video shows serial images acquired from 200 to 500 μm below the organoid surface. (AVI 4165 kb)

Active blood flow within the grafted organoid.

Two-photon time series of grafted organoid after dextran injection at 120 dpi showing active blood flow at 141 μm depth. (MP4 18013 kb)

In vivo two-photon imaging of organoid tissue labeled with jRGECO1a, a genetically encoded calcium sensor.

This representative 60 s movie of calcium activity was acquired at 78 dpi. The mouse was head-fixed to the microscope but unanesthetized and allowed to run on a cylindrical treadmill. (AVI 3980 kb)

In vivo two-photon imaging of organoid tissue labeled with jRGECO1a.

This representative 60 s movie of calcium activity was acquired at 108 dpi. The mouse was head-fixed to the microscope but unanesthetized and allowed to run on a cylindrical treadmill. (AVI 2567 kb)

Additional example of in vivo two-photon imaging of organoid tissue labeled with jRGECO1a.

This representative 60 s movie of calcium activity was acquired at 109 dpi. (AVI 2266 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Mansour, A., Gonçalves, J., Bloyd, C. et al. An in vivo model of functional and vascularized human brain organoids. Nat Biotechnol 36, 432–441 (2018). https://doi.org/10.1038/nbt.4127

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nbt.4127

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