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:

A 3D human neural cell culture system for modeling Alzheimer's disease

This article has been updated

Abstract

Stem cell technologies have facilitated the development of human cellular disease models that can be used to study pathogenesis and test therapeutic candidates. These models hold promise for complex neurological diseases such as Alzheimer's disease (AD), because existing animal models have been unable to fully recapitulate all aspects of pathology. We recently reported the characterization of a novel 3D culture system that exhibits key events in AD pathogenesis, including extracellular aggregation of amyloid-β (Aβ) and accumulation of hyperphosphorylated tau. Here we provide instructions for the generation and analysis of 3D human neural cell cultures, including the production of genetically modified human neural progenitor cells (hNPCs) with familial AD mutations, the differentiation of the hNPCs in a 3D matrix and the analysis of AD pathogenesis. The 3D culture generation takes 1–2 d. The aggregation of Aβ is observed after 6 weeks of differentiation, followed by robust tau pathology after 10–14 weeks.

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: Polycistronic lentiviral vectors used in this study.
Figure 2: Overview of the ReN VM cell 3D culture protocol.
Figure 3: FACS enrichment of ReN-G and ReN-GA cells for higher expressions of APP with FAD mutations.
Figure 4: Various 3D culture formats.
Figure 5: Analysis of soluble Aβ levels in the enriched FAD ReN cells by western blotting and Aβ ELISA.
Figure 6: Images of ReN-G2-differentiated neural cell populations in 3D cultures after 2- and 4-week differentiation.
Figure 7: Reconstitution of Aβ aggregates in 3D-cultured FAD ReN cells.
Figure 8: p-Tau accumulation in cell body and neurites of the enriched ReN-mAP cells.
Figure 9: Detection of aggregated p-tau in the enriched ReN-G and HReN-mGAP cells.

Similar content being viewed by others

Change history

  • 01 July 2015

     In the version of this article initially published online, the y-axis labels in Figure 5b–d were incorrectly given as pmol/ml. The correct label is pmol/liter. The error has been corrected for the print, PDF and HTML versions of this article.

References

  1. Tanzi, R.E. & Bertram, L. Twenty years of the Alzheimer's disease amyloid hypothesis: a genetic perspective. Cell 120, 545–555 (2005).

    Article  CAS  PubMed  Google Scholar 

  2. Götz, J. & Ittner, L.M. Animal models of Alzheimer's disease and frontotemporal dementia. Nat. Rev. Neurosci. 9, 532–544 (2008).

    Article  PubMed  Google Scholar 

  3. Chin, J. Selecting a mouse model of Alzheimer's disease. Methods Mol. Biol. 670, 169–189 (2011).

    Article  CAS  PubMed  Google Scholar 

  4. Oddo, S. et al. Triple-transgenic model of Alzheimer's disease with plaques and tangles: intracellular Aβ and synaptic dysfunction. Neuron 39, 409–421 (2003).

    Article  CAS  PubMed  Google Scholar 

  5. Yagi, T. et al. Modeling familial Alzheimer's disease with induced pluripotent stem cells. Hum. Mol. Genet. 20, 4530–4539 (2011).

    Article  CAS  PubMed  Google Scholar 

  6. Israel, M.A. et al. Probing sporadic and familial Alzheimer's disease using induced pluripotent stem cells. Nature 482, 216–220 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Shi, Y. et al. A human stem cell model of early Alzheimer's disease pathology in Down syndrome. Sci. Transl. Med. 4, 124ra29 (2012).

    PubMed  PubMed Central  Google Scholar 

  8. Kondo, T. et al. Modeling Alzheimer's disease with iPSCs reveals stress phenotypes associated with intracellular Aβ and differential drug responsiveness. Cell Stem Cell 12, 487–496 (2013).

    Article  CAS  PubMed  Google Scholar 

  9. Sproul, A.A. et al. Characterization and molecular profiling of PSEN1 familial Alzheimer's disease iPSC-derived neural progenitors. PLoS ONE 9, e84547 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  10. Muratore, C.R. et al. The familial Alzheimer's disease APPV717I mutation alters APP processing and tau expression in iPSC-derived neurons. Hum. Mol. Genet. 23, 3523–3536 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Choi, S.H. & Tanzi, R.E. iPSCs to the rescue in Alzheimer's research. Cell Stem Cell 10, 235–236 (2012).

    Article  CAS  PubMed  Google Scholar 

  12. Saha, K. & Jaenisch, R. Technical challenges in using human induced pluripotent stem cells to model disease. Cell Stem Cell 5, 584–595 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Young, J.E. & Goldstein, L.S.B. Alzheimer's disease in a dish: promises and challenges of human stem cell models. Hum. Mol. Genet. 21, R82–R89 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Choi, S.H. et al. A three-dimensional human neural cell culture model of Alzheimer's disease. Nature 515, 274–278 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Donato, R. et al. Differential development of neuronal physiological responsiveness in two human neural stem cell lines. BMC Neurosci. 8, 36 (2007).

    Article  PubMed  PubMed Central  Google Scholar 

  16. Hoffrogge, R. et al. 2-DE proteome analysis of a proliferating and differentiating human neuronal stem cell line (ReNcell VM). Proteomics 6, 1833–1847 (2006).

    Article  CAS  PubMed  Google Scholar 

  17. Beyer, S. et al. Neuroproteomics in stem cell differentiation. Proteomics Clin. Appl. 1, 1513–1523 (2007).

    Article  CAS  PubMed  Google Scholar 

  18. Le, M.T.N. et al. MicroRNA-125b promotes neuronal differentiation in human cells by repressing multiple targets. Mol. Cell Biol. 29, 5290–5305 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Morgan, P.J. et al. Protection of neurons derived from human neural progenitor cells by veratridine. Neuroreport 20, 1225–1229 (2009).

    Article  CAS  PubMed  Google Scholar 

  20. Lange, C. et al. Small molecule GSK-3 inhibitors increase neurogenesis of human neural progenitor cells. Neurosci. Lett. 488, 36–40 (2011).

    Article  CAS  PubMed  Google Scholar 

  21. Hernández-Benítez, R., Vangipuram, S.D., Ramos-Mandujano, G., Lyman, W.D. & Pasantes-Morales, H. Taurine enhances the growth of neural precursors derived from fetal human brain and promotes neuronal specification. Dev. Neurosci. 35, 40–49 (2013).

    Article  PubMed  Google Scholar 

  22. Mazemondet, O. et al. Quantitative and kinetic profile of Wnt/β-catenin signaling components during human neural progenitor cell differentiation. Cell. Mol. Biol. Lett. 16, 515–538 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Liedmann, A., Rolfs, A. & Frech, M.J. Cultivation of human neural progenitor cells in a 3-dimensional self-assembling peptide hydrogel. J. Vis. Exp 59, e3830 (2012).

    Google Scholar 

  24. Hovakimyan, M. et al. Survival of transplanted human neural stem cell line (ReNcell VM) into the rat brain with and without immunosuppression. Ann. Anat. 194, 429–435 (2012).

    Article  CAS  PubMed  Google Scholar 

  25. Hübner, R. et al. Differentiation of human neural progenitor cells regulated by Wnt-3a. Biochem. Biophys. Res. Commun. 400, 358–362 (2010).

    Article  PubMed  Google Scholar 

  26. Jaeger, A., Baake, J., Weiss, D.G. & Kriehuber, R. Glycogen synthase kinase-3β regulates differentiation-induced apoptosis of human neural progenitor cells. Int. J. Dev. Neurosci. 31, 61–68 (2013).

    Article  CAS  PubMed  Google Scholar 

  27. Chaudhry, Z.L. & Ahmed, B.Y. Caspase-2 and caspase-8 trigger caspase-3 activation following 6-OHDA-induced stress in human dopaminergic neurons differentiated from ReNVM stem cells. Neurol. Res. 35, 435–440 (2013).

    Article  CAS  PubMed  Google Scholar 

  28. Hughes, C.S., Postovit, L.M. & Lajoie, G.A. Matrigel: a complex protein mixture required for optimal growth of cell culture. Proteomics 10, 1886–1890 (2010).

    Article  CAS  PubMed  Google Scholar 

  29. Ortinau, S. et al. Effect of 3D-scaffold formation on differentiation and survival in human neural progenitor cells. Biomed. Eng. Online 9, 70 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  30. LaPlaca, M.C., Vernekar, V.N., Shoemaker, J.T. & Cullen, D.K. Three-dimensional neuronal cultures. in Methods in Bioengineering: 3D Tissue Engineering (eds. Berthiaume, F. & Morgan, J.) 187–204 (Artech House 2010).

  31. Suga, H. et al. Self-formation of functional adenohypophysis in three-dimensional culture. Nature 480, 57–62 (2011).

    Article  CAS  PubMed  Google Scholar 

  32. Li, H., Wijekoon, A. & Leipzig, N.D. 3D differentiation of neural stem cells in macroporous photopolymerizable hydrogel scaffolds. PLoS ONE 7, e48824 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Liedmann, A., Frech, S., Morgan, P.J., Rolfs, A. & Frech, M.J. Differentiation of human neural progenitor cells in functionalized hydrogel matrices. Biores. Open Access 1, 16–24 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Tang-Schomer, M.D. et al. Bioengineered functional brain-like cortical tissue. Proc. Natl. Acad. Sci. USA 111, 13811–13816 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Falk, A. et al. Capture of neuroepithelial-like stem cells from pluripotent stem cells provides a versatile system for in vitro production of human neurons. PLoS ONE 7, e29597 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Zhang, D. et al. A 3D Alzheimer's disease culture model and the induction of P21-activated kinase mediated sensing in iPSC derived neurons. Biomaterials 35, 1420–1428 (2014).

    Article  CAS  PubMed  Google Scholar 

  37. Cathomen, T. & Joung, J.K. Zinc-finger nucleases: the next generation emerges. Mol. Ther. 16, 1200–1207 (2008).

    Article  CAS  PubMed  Google Scholar 

  38. Mussolino, C. & Cathomen, T. TALE nucleases: tailored genome engineering made easy. Curr. Opin. Biotechnol. 23, 644–650 (2012).

    Article  CAS  PubMed  Google Scholar 

  39. Hsu, P.D., Lander, E.S. & Zhang, F. Development and applications of CRISPR-Cas9 for genome engineering. Cell 157, 1262–1278 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Iovino, M., Patani, R., Watts, C., Chandran, S. & Spillantini, M.G. Human stem cell-derived neurons: a system to study human tau function and dysfunction. PLoS ONE 5, e13947 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  41. Ingelsson, M. et al. No alteration in tau exon 10 alternative splicing in tangle-bearing neurons of the Alzheimer's disease brain. Acta Neuropathol. 112, 439–449 (2006).

    Article  CAS  PubMed  Google Scholar 

  42. Sena-Esteves, M., Tebbets, J.C., Steffens, S., Crombleholme, T. & Flake, A.W. Optimized large-scale production of high titer lentivirus vector pseudotypes. J. Virol. Methods 122, 131–139 (2004).

    Article  CAS  PubMed  Google Scholar 

  43. Livak, K.J. & Schmittgen, T.D. Analysis of relative gene expression data using real-time quantitative PCR and the 2(–ΔΔC(T)) method. Methods 25, 402–408 (2001).

    Article  CAS  PubMed  Google Scholar 

  44. Schmued, L. et al. Introducing Amylo-Glo, a novel fluorescent amyloid specific histochemical tracer especially suited for multiple labeling and large scale quantification studies. J. Neurosci. Methods 209, 120–126 (2012).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work is supported by grants from the Cure Alzheimer's Fund to D.Y.K. and R.E.T., from the US National Institutes of Health (NIH) 1RF1AG048080-01 (D.Y.K. and R.E.T.), 5P01AG15379 (D.Y.K. and R.E.T.), 2R01AG014713 (D.Y.K.) and 5R37MH060009 (R.E.T.), from the German Federal Ministry for Education and Research BioPharma-NeuroAllianz 0315608A (O.B.), from EU SCR&Tox HEALTH-F5-2010-266753 (O.B.), and from Bio & Medical Technology Development Program of the National Research Foundation (funded by the Korean government) (Y.H.K.). We appreciate B.T. Hyman, O. Berezovska, D.M. Kovacs (MGH), J. Hardy (NIH) and P. Davies (Albert Einstein College of Medicine) for providing cDNAs and antibodies. We also acknowledge B.A. Tannos at MGH Viral Vector Core (supported by NIH/National Institute of Neurological Disorders and Stroke P30NS04776), M. Waring at Ragon Institute's Imaging Core facility (part of the Harvard Center for AIDS Research Immunology Core), and M.L. McKee and D. Capen at MGH Microscopy Core of the Center for Systems Biology for providing the detailed protocols and for revising the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

D.Y.K. and R.E.T. were equally responsible for experimental design and supervising the whole project. Y.H.K., S.H.C., C.D. and D.Y.K. mainly contributed to writing and revising the manuscript. C.D., E.B., K.J.W., M.H. and D.Y.K. performed the experiments. M.H., O.B., J.B.K. and C.S. contribute to writing the manuscript.

Corresponding authors

Correspondence to Rudolph E Tanzi or Doo Yeon Kim.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Pictures showing each steps of our 3D culture protocol.

The numbers in each picturematch with those of 3D culture steps described within the protocol.

Supplementary Figure 2 Extracellular Aβ deposits increased in 3D-differentiated ReN VM cells withFAD mutations.

a. Detection of amyloid plaques with Amylo-Glo, a fluorescent amyloidspecificdye, in control ReN-G and FAD HReN-mGAP cells 3D-differentiated for 7 and 17weeks. HReN-mGAP cells, ReN-G cells were transfected with APPSL/PSΔE9/mCherryvectors, enriched with FACS to achieve high Aβ secretion; Blue, Amylo-Glo; Red,Propidium Iodide (P.I.); Scale bar, 50 μm. b. The number and size of Amylo-Glo-stainedamyloid structures were increased in 17-week differentiated FAD HReN-mGAP cells ascompared to 7-week 3D cultures (Blue, Amylo-Glo).

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–2 (PDF 466 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kim, Y., Choi, S., D'Avanzo, C. et al. A 3D human neural cell culture system for modeling Alzheimer's disease. Nat Protoc 10, 985–1006 (2015). https://doi.org/10.1038/nprot.2015.065

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nprot.2015.065

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