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:

Modeling amyloid beta and tau pathology in human cerebral organoids

Abstract

The typical abnormalities observed in the brain of Alzheimer’s disease (AD) patients include synaptic alterations, neuronal death, brain inflammation, and the accumulation of protein aggregates in the form of amyloid plaques and neurofibrillary tangles. Despite the development of many animal and in vitro models for AD, there is a lack of an experimental approach that fully recapitulates essential aspects of the disease in human cells. Here, we report the generation of a new model to study AD, consisting of cerebral organoids (COs) produced from human-induced pluripotent stem cells (iPSCs). Under our experimental conditions, COs grow to form three-dimensional (3D) structures containing neural areas with cortical-like organization. Analysis of COs by histological and biochemical methods revealed that organoids produced from iPSCs derived from patients affected by familial AD or Down syndrome (DS) spontaneously develop over time pathological features of AD, including accumulation of structures highly reminiscent to amyloid plaques and neurofibrillary tangles. These pathological abnormalities were not observed in COs generated from various controls, including human iPSCs from healthy individuals, human iPSCs from patients affected by Creutzfeldt–Jakob disease, mouse embryonic stem cells (ESCs), or mouse iPSCs. These findings enable modeling genetic AD in a human cellular context in a 3D cortical-like tissue developed in vitro from patient-specific stem cells. This system provides a more relevant disease model compared to pre-existing methods and offers a new platform for discovery of novel targets and screening of drugs for therapeutic intervention.

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
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7
Fig. 8

Similar content being viewed by others

References

  1. Querfurth HW, LaFerla FM. Alzheimer’s disease. N Engl J Med. 2010;362:329–44.

    Article  CAS  Google Scholar 

  2. Laurijssens B, Aujard F, Rahman A. Animal models of Alzheimer’s disease and drug development. Drug Discov Today Technol. 2013;10:e319–27.

    Article  Google Scholar 

  3. Duyckaerts C, Potier MC, Delatour B. Alzheimer disease models and human neuropathology: similarities and differences. Acta Neuropathol. 2008;115:5–38.

    Article  Google Scholar 

  4. Stadtfeld M, Hochedlinger K. Induced pluripotency: history, mechanisms, and applications. Genes Dev. 2010;24:2239–63.

    Article  CAS  Google Scholar 

  5. Yagi T, Ito D, Okada Y, Akamatsu W, Nihei Y, Yoshizaki T. et al. Modeling familial Alzheimer’s disease with induced pluripotent stem cells. Hum Mol Genet. 2011;20:4530–9.

    Article  CAS  Google Scholar 

  6. Israel MA, Yuan SH, Bardy C, Reyna SM, Mu Y, Herrera C. et al. Probing sporadic and familial Alzheimer’s disease using induced pluripotent stem cells. Nature. 2012;482:216–20.

    Article  CAS  Google Scholar 

  7. Kondo T, Asai M, Tsukita K, Kutoku Y, Ohsawa Y, Sunada Y. et al. Modeling Alzheimer’s disease with iPSCs reveals stress phenotypes associated with intracellular Abeta and differential drug responsiveness. Cell Stem Cell. 2013;12:487–96.

    Article  CAS  Google Scholar 

  8. Shi Y, Kirwan P, Smith J, MacLean G, Orkin SH, Livesey FJ. A human stem cell model of early Alzheimer’s disease pathology in Down syndrome. Sci Transl Med. 2012;4:124ra29

    PubMed  PubMed Central  Google Scholar 

  9. Muratore CR, Rice HC, Srikanth P, Callahan DG, Shin T, Benjamin LN. et al. The familial Alzheimer’s disease APPV717I mutation alters APP processing and Tau expression in iPSC-derived neurons. Hum Mol Genet. 2014;23:3523–36.

    Article  CAS  Google Scholar 

  10. Moore S, Evans LD, Andersson T, Portelius E, Smith J, Dias TB. et al. APP metabolism regulates tau proteostasis in human cerebral cortex neurons. Cell Rep. 2015;11:689–96.

    Article  CAS  Google Scholar 

  11. Koch P, Tamboli IY, Mertens J, Wunderlich P, Ladewig J, Stuber K. et al. Presenilin-1 L166P mutant human pluripotent stem cell-derived neurons exhibit partial loss of gamma-secretase activity in endogenous amyloid-beta generation. Am J Pathol. 2012;180:2404–16.

    Article  CAS  Google Scholar 

  12. Sproul AA, Jacob S, Pre D, Kim SH, Nestor MW, Navarro-Sobrino M. et al. Characterization and molecular profiling of PSEN1 familial Alzheimer’s disease iPSC-derived neural progenitors. PLoS ONE. 2014;9:e84547

    Article  Google Scholar 

  13. Mahairaki V, Ryu J, Peters A, Chang Q, Li T, Park TS. et al. Induced pluripotent stem cells from familial Alzheimer’s disease patients differentiate into mature neurons with amyloidogenic properties. Stem Cells Dev. 2014;23:2996–3010.

    Article  CAS  Google Scholar 

  14. Armijo E, Gonzalez C, Shahnawaz M, Flores A, Davis B, Soto C. Increased susceptibility to Abeta toxicity in neuronal cultures derived from familial Alzheimer’s disease (PSEN1-A246E) induced pluripotent stem cells. Neurosci Lett. 2017;639:74–81.

    Article  CAS  Google Scholar 

  15. Lancaster MA, Renner M, Martin CA, Wenzel D, Bicknell LS, Hurles ME. et al. Cerebral organoids model human brain development and microcephaly. Nature. 2013;501:373–9.

    Article  CAS  Google Scholar 

  16. Lancaster MA, Knoblich JA. Generation of cerebral organoids from human pluripotent stem cells. Nat Protoc. 2014;9:2329–40.

    Article  CAS  Google Scholar 

  17. Sherrington R, Rogaev EI, Liang Y, Rogaeva EA, Levesque G, Ikeda M. et al. Cloning of a gene bearing missense mutations in early-onset familial Alzheimer’s disease. Nature. 1995;375:754–60.

    Article  CAS  Google Scholar 

  18. Nethercott HE, Brick DJ, Schwartz PH. Derivation of induced pluripotent stem cells by lentiviral transduction. Methods Mol Biol. 2011;767:67–85.

    Article  CAS  Google Scholar 

  19. Liu Y, Zheng Y, Li S, Xue H, Schmitt K, Hergenroeder GW. et al. Human neural progenitors derived from integration-free iPSCs for SCI therapy. Stem Cell Res. 2017;19:55–64.

    Article  CAS  Google Scholar 

  20. Renner M, Lancaster MA, Bian S, Choi H, Ku T, Peer A. et al. Self-organized developmental patterning and differentiation in cerebral organoids. EMBO J. 2017;36:1316–29.

    Article  CAS  Google Scholar 

  21. Klunk WE, Wang Y, Huang GF, Debnath ML, Holt DP, Mathis CA. Uncharged thioflavin-T derivatives bind to amyloid-beta protein with high affinity and readily enter the brain. Life Sci. 2001;69:1471–84.

    Article  CAS  Google Scholar 

  22. Kawarabayashi T, Younkin LH, Saido TC, Shoji M, Ashe KH, Younkin SG. Age-dependent changes in brain, CSF, and plasma amyloid (beta) protein in the Tg2576 transgenic mouse model of Alzheimer’s disease. J Neurosci. 2001;21:372–81.

    Article  CAS  Google Scholar 

  23. Rostagno A, Ghiso J. Isolation and biochemical characterization of amyloid plaques and paired helical filaments. Curr Protoc Cell Biol. 2009;Chapter 3:Unit 3.33.

  24. Porter AG, Janicke RU. Emerging roles of caspase-3 in apoptosis. Cell Death Differ. 1999;6:99–104.

    Article  CAS  Google Scholar 

  25. Zhang D, Pekkanen-Mattila M, Shahsavani M, Falk A, Teixeira AI, Herland A. A 3D Alzheimer’s disease culture model and the induction of P21-activated kinase mediated sensing in iPSC derived neurons. Biomaterials. 2014;35:1420–8.

    Article  CAS  Google Scholar 

  26. Choi SH, Kim YH, Hebisch M, Sliwinski C, Lee S, D’Avanzo C. et al. A three-dimensional human neural cell culture model of Alzheimer’s disease. Nature. 2014;515:274–8.

    Article  CAS  Google Scholar 

  27. Raja WK, Mungenast AE, Lin YT, Ko T, Abdurrob F, Seo J. et al. Self-organizing 3D human neural tissue derived from induced pluripotent stem cells recapitulate Alzheimer’s disease phenotypes. PLoS ONE. 2016;11:e0161969

    Article  Google Scholar 

  28. Garcez PP, Loiola EC, Madeiro da CR, Higa LM, Trindade P, Delvecchio R. et al. Zika virus impairs growth in human neurospheres and brain organoids. Science. 2016;352:816–8.

    Article  CAS  Google Scholar 

  29. Huch M, Knoblich JA, Lutolf MP, Martinez-Arias A. The hope and the hype of organoid research. Development. 2017;144:938–41.

    Article  CAS  Google Scholar 

  30. Musiek ES, Holtzman DM. Three dimensions of the amyloid hypothesis: time, space and ‘wingmen’. Nat Neurosci. 2015;18:800–6.

    Article  CAS  Google Scholar 

  31. Lancaster MA, Knoblich JA. Organogenesis in a dish: modeling development and disease using organoid technologies. Science. 2014;345:1247125

    Article  Google Scholar 

  32. Qian X, Nguyen HN, Song MM, Hadiono C, Ogden SC, Hammack C. et al. Brain-region-specific organoids using mini-bioreactors for modeling ZIKV exposure. Cell. 2016;165:1238–54.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We are grateful to Dr. Brian Davis (University of Texas Medical School at Houston) for his valuable help in establishing the iPSC technology in our lab, Dr. Fabrizio Tagliavini (Istituto Carlo Besta, Italy) for kindly providing human fibroblasts from patients affected by inherited Creutzfeldt–Jakob disease. We are also grateful to Dr. Ying Liu (Institute of Molecular Medicine, University of Texas at Houston) for providing an iPSC line obtained from healthy controls. This project was partially funded by a pilot grant from the University of Texas Brain Initiative Program.

Author contributions

CG designed the studies, carried out many of the experiments, analyzed the results, and prepared the first version of the figures. EA generated and characterized induced pluripotent stem cells used in these studies, performed some of the experiments and histological staining, analyzed some of the results, prepared the final version of the figures, and assisted in the preparation and writing of the final version of the manuscript. JB-A performed the biochemical studies to measure the levels of Aβ and tau aggregates. AB-C participated in the studies with mouse cerebral organoids. CM performed the western blot and ELISA studies of p-tau, generated induced pluripotent stem cells, produced the cerebral organoids derived from Creutzfeldt–Jakob disease cells, processed tissue samples, and assisted in the preparation and writing of the final version of the manuscript. CS is the principal investigator on the project and was responsible for coordinating research activity, analyzing the data, funding, writing the manuscript, and producing the final version of the article.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Claudio Soto.

Ethics declarations

Conflict of interest

The authors declare that they have no conflict of interest.

Electronic supplementary material

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Gonzalez, C., Armijo, E., Bravo-Alegria, J. et al. Modeling amyloid beta and tau pathology in human cerebral organoids. Mol Psychiatry 23, 2363–2374 (2018). https://doi.org/10.1038/s41380-018-0229-8

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41380-018-0229-8

This article is cited by

Search

Quick links