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.

  • Letter
  • Published:

Probing sporadic and familial Alzheimer’s disease using induced pluripotent stem cells

This article has been updated

Abstract

Our understanding of Alzheimer’s disease pathogenesis is currently limited by difficulties in obtaining live neurons from patients and the inability to model the sporadic form of the disease. It may be possible to overcome these challenges by reprogramming primary cells from patients into induced pluripotent stem cells (iPSCs). Here we reprogrammed primary fibroblasts from two patients with familial Alzheimer’s disease, both caused by a duplication of the amyloid-β precursor protein gene1 (APP; termed APPDp), two with sporadic Alzheimer’s disease (termed sAD1, sAD2) and two non-demented control individuals into iPSC lines. Neurons from differentiated cultures were purified with fluorescence-activated cell sorting and characterized. Purified cultures contained more than 90% neurons, clustered with fetal brain messenger RNA samples by microarray criteria, and could form functional synaptic contacts. Virtually all cells exhibited normal electrophysiological activity. Relative to controls, iPSC-derived, purified neurons from the two APPDp patients and patient sAD2 exhibited significantly higher levels of the pathological markers amyloid-β(1–40), phospho-tau(Thr 231) and active glycogen synthase kinase-3β (aGSK-3β). Neurons from APPDp and sAD2 patients also accumulated large RAB5-positive early endosomes compared to controls. Treatment of purified neurons with β-secretase inhibitors, but not γ-secretase inhibitors, caused significant reductions in phospho-Tau(Thr 231) and aGSK-3β levels. These results suggest a direct relationship between APP proteolytic processing, but not amyloid-β, in GSK-3β activation and tau phosphorylation in human neurons. Additionally, we observed that neurons with the genome of one sAD patient exhibited the phenotypes seen in familial Alzheimer’s disease samples. More generally, we demonstrate that iPSC technology can be used to observe phenotypes relevant to Alzheimer’s disease, even though it can take decades for overt disease to manifest in patients.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Generation of iPSC lines and purified neurons from APP Dp , sAD and NDC fibroblasts.
Figure 2: Increased amyloid-β, p-tau and aGSK-3β in sAD2 and APP Dp neuronal cultures.
Figure 3: Analysis of early endosome and synapsin levels in purified neurons co-cultured with astrocytes.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

Data deposits

Data have been deposited in the Gene ExpressionOmnibus under accession GSE34879.

Change history

  • 08 February 2012

    An addition was made to Acknowlegements.

References

  1. Rovelet-Lecrux, A. et al. APP locus duplication in a Finnish family with dementia and intracerebral haemorrhage. J. Neurol. Neurosurg. Psychiatry 78, 1158–1159 (2007)

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  3. Ballatore, C., Lee, V. M. Y. & Trojanowski, J. Q. Tau-mediated neurodegeneration in Alzheimer’s disease and related disorders. Nature Rev. Neurosci. 8, 663–672 (2007)

    Article  CAS  Google Scholar 

  4. Gatz, M. et al. Role of genes and environments for explaining Alzheimer disease. Arch. Gen. Psychiatry 63, 168–174 (2006)

    Article  Google Scholar 

  5. Games, D. et al. Alzheimer-type neuropathology in transgenic mice overexpressing V717F β-amyloid precursor protein. Nature 373, 523–527 (1995)

    Article  ADS  CAS  Google Scholar 

  6. Roberson, E. D. et al. Reducing endogenous tau ameliorates amyloid β-induced deficits in an Alzheimer’s disease mouse model. Science 316, 750–754 (2007)

    Article  ADS  CAS  Google Scholar 

  7. Busciglio, J., Lorenzo, A., Yeh, J. & Yankner, B. A. β-Amyloid fibrils induce tau phosphorylation and loss of microtubule binding. Neuron 14, 879–888 (1995)

    Article  CAS  Google Scholar 

  8. Ebert, A. D. et al. Induced pluripotent stem cells from a spinal muscular atrophy patient. Nature 457, 277–280 (2009)

    Article  ADS  CAS  Google Scholar 

  9. Nguyen, H. N. et al. LRRK2 mutant iPSC-derived DA neurons demonstrate increased susceptibility to oxidative stress. Cell Stem Cell 8, 267–280 (2011)

    Article  CAS  Google Scholar 

  10. Qiang, L. et al. Directed conversion of Alzheimer’s disease patient skin fibroblasts into functional neurons. Cell 3, 359–371 (2011)

    Article  Google Scholar 

  11. Urbach, A., Bar-Nur, O., Daley, G. Q. & Benvenisty, N. Differential modeling of fragile X syndrome by human embryonic stem cells and induced pluripotent stem cells. Cell Stem Cell 6, 407–411 (2010)

    Article  CAS  Google Scholar 

  12. Yuan, S. H. et al. Cell-surface marker signatures for the isolation of neural stem cells, glia and neurons derived from human pluripotent stem cells. PLoS ONE 6, e17540 (2011)

    Article  ADS  CAS  Google Scholar 

  13. Cao, X., Pfaff, S. L. & Gage, F. H. YAP regulates neural progenitor cell number via the TEA domain transcription factor. Genes Dev. 22, 3320–3334 (2008)

    Article  CAS  Google Scholar 

  14. Reddy, B. V. & Irvine, K. D. Regulation of Drosophila glial cell proliferation by Merlin-Hippo signaling. Development 138, 5201–5212 (2011)

    Article  CAS  Google Scholar 

  15. Citron, M. et al. Excessive production of amyloid β-protein by peripheral cells of symptomatic and presymptomatic patients carrying the Swedish familial Alzheimer disease mutation. Proc. Natl Acad. Sci. USA 91, 11993–11997 (1994)

    Article  ADS  CAS  Google Scholar 

  16. Scheuner, D. et al. Secreted amyloid β-protein similar to that in the senile plaques of Alzheimer’s disease is increased in vivo by the presenilin 1 and 2 and APP mutations linked to familial Alzheimer’s disease. Nature Med. 2, 864–870 (1996)

    Article  CAS  Google Scholar 

  17. Gasparini, L. et al. Peripheral markers in testing pathophysiological hypotheses and diagnosing Alzheimer’s disease. FASEB J. 12, 17–34 (1998)

    Article  CAS  Google Scholar 

  18. Arriagada, P. V., Growdon, J. H., Hedley-Whyte, E. T. & Hyman, B. T. Neurofibrillary tangles but not senile plaques parallel duration and severity of Alzheimer’s disease. Neurology 42, 631 (1992)

    Article  CAS  Google Scholar 

  19. Cho, J.-H. & Johnson, G. V. W. Primed phosphorylation of tau at Thr 231 by glycogen synthase kinase 3β (GSK3β) plays a critical role in regulating tau’s ability to bind and stabilize microtubules. J. Neurochem. 88, 349–358 (2004)

    Article  CAS  Google Scholar 

  20. Buerger, K. et al. CSF tau protein phosphorylated at threonine 231 correlates with cognitive decline in MCI subjects. Neurology 59, 627–629 (2002)

    Article  CAS  Google Scholar 

  21. Buerger, K. et al. CSF phosphorylated tau protein correlates with neocortical neurofibrillary pathology in Alzheimer’s disease. Brain 129, 3035–3041 (2006)

    Article  Google Scholar 

  22. Cho, J. & Johnson, G. Glycogen synthase kinase 3β phosphorylates tau at both primed and unprimed sites. J. Biol. Chem. 278, 187–193 (2003)

    Article  CAS  Google Scholar 

  23. Dajani, R. et al. Crystal structure of glycogen synthase kinase 3β: structural basis for phosphate-primed substrate specificity and autoinhibition. Cell 105, 721–732 (2001)

    Article  CAS  Google Scholar 

  24. Cataldo, A. M. et al. Endocytic pathway abnormalities precede amyloid β deposition in sporadic Alzheimer’s disease and Down syndrome: differential effects of APOE genotype and presenilin mutations. Am. J. Pathol. 157, 277–286 (2000)

    Article  CAS  Google Scholar 

  25. Cataldo, A. et al. Endocytic disturbances distinguish among subtypes of Alzheimer’s disease and related disorders. Ann. Neurol. 50, 661–665 (2001)

    Article  CAS  Google Scholar 

  26. Nixon, R. A. Endosome function and dysfunction in Alzheimer’s disease and other neurodegenerative diseases. Neurobiol. Aging 26, 373–382 (2005)

    Article  CAS  Google Scholar 

  27. Hamos, J. E., DeGennaro, L. J. & Drachman, D. A. Synaptic loss in Alzheimer’s disease and other dementias. Neurology 39, 355–361 (1989)

    Article  CAS  Google Scholar 

  28. Qin, S., Hu, X. Y., Xu, H. & Zhou, J. N. Regional alteration of synapsin I in the hippocampal formation of Alzheimer’s disease patients. Acta Neuropathol. 107, 209–215 (2004)

    Article  Google Scholar 

  29. Salehi, A. et al. Increased App expression in a mouse model of Down’s syndrome disrupts NGF transport and causes cholinergic neuron degeneration. Neuron 51, 29–42 (2006)

    Article  CAS  Google Scholar 

  30. Jiang, Y. et al. Alzheimer’s-related endosome dysfunction in Down syndrome is Aβ-independent but requires APP and is reversed by BACE-1 inhibition. Proc. Natl Acad. Sci. USA 107, 1630–1635 (2010)

    Article  ADS  CAS  Google Scholar 

  31. Takashima, A. in Current Protocols in Cell Biology Ch. 2 12 (John Wiley & Sons, 2001)

    Google Scholar 

  32. Park, I.-H., Lerou, P. H., Zhao, R., Huo, H. & Daley, G. Q. Generation of human-induced pluripotent stem cells. Nature Protocols 3, 1180–1186 (2008)

    Article  CAS  Google Scholar 

  33. Akagi, T., Sasai, K. & Hanafusa, H. Refractory nature of normal human diploid fibroblasts with respect to oncogene-mediated transformation. Proc. Natl Acad. Sci. USA 100, 13567–13572 (2003)

    Article  ADS  CAS  Google Scholar 

  34. Emre, N. et al. The ROCK inhibitor Y-27632 improves recovery of human embryonic stem cells after fluorescence-activated cell sorting with multiple cell surface markers. PLoS ONE 5, e12148 (2010)

    Article  ADS  Google Scholar 

  35. Takahashi, K. et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 131, 861–872 (2007)

    Article  CAS  Google Scholar 

  36. Kawasaki, H. et al. Induction of midbrain dopaminergic neurons from ES cells by stromal cell derived inducing activity. Neuron 28, 31–40 (2000)

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank D. Galasko, M. Sundsmo, J. Rivera, J. Fontaine, C. Gigliotti and B. Yu at the University of California, San Diego (UCSD) Alzheimer’s Disease Research Center for patient samples and data (grant AGO 5131); S. Dowdy and N. Yoshioka for viral vectors; B. Balderas at BD Biosciences for antibodies; C. Santucci and S. Nguyen for teratoma assay assistance; the UCSD Neuroscience Microscopy Shared Facility (grant P30 NS047101); and Planned Parenthood of the Pacific Southwest for fetal brain specimens. Funding was from California Institute of Regenerative Medicine (CIRM) comprehensive grants (M.M., F.H.G., L.S.B.G.), CIRM predoctoral fellowship (M.A.I.), FP7 Marie Curie IOF (C.B.), Weatherstone Foundation fellowship (K.L.N.), National Institutes of Health K12 HD001259, the Hartwell Foundation (L.C.L., F.S.B.), the Lookout Fund and the McDonnell Foundation (F.H.G.). L.S.B.G. is an investigator with the Howard Hughes Medical Institute. Some experiments were conducted in J.F. Loring's laboratory (The Scripps Research Institute) with support from grants TR1-01250, CL1-00502, RM1-01717 (CIRM) and a gift from the Esther O'Keefe Foundation.

Author information

Authors and Affiliations

Authors

Contributions

M.A.I. and L.S.B.G. conceived the project; M.A.I. and L.S.B.G. designed the experiments; M.A.I., S.H.Y., C.B., S.M.R., Y.M., C.H., M.P.H., S.V.G., M.M., K.L.N. and F.S.B. performed the experiments; M.A.I., S.H.Y. and C.T.C. developed differentiation methods; A.M.R. and E.H.K. provided APPDp patient samples and information; F.H.G. supervised C.B. and Y.M.; M.M. supervised M.P.H. and S.V.G.; L.C.L. supervised K.L.N. and F.S.B.; M.A.I. and L.S.B.G. wrote the manuscript; F.H.G., E.H.K. and A.M.R. edited the manuscript.

Corresponding author

Correspondence to Lawrence S. B. Goldstein.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Information

This file contains Supplementary Figures 1-12 with legends and Supplementary Tables 1, 3 and 4 (see separate files for Supplementary Tables 2 and 5). (PDF 11256 kb)

Supplementary Table 2

This table shows differentially expressed genes between purified neurons and fetal brain versus non-neural fetal samples (sheet 1) and differentially expressed genes between purified neurons versus fetal brain (sheet 2). (XLS 2984 kb)

Supplementary Table 5

This table contains Amyloid-β, aGSK3β and p-tau/total tau detailed by patient, iPSC line and culture well. (PDF 225 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Cite this article

Israel, M., Yuan, S., Bardy, C. et al. Probing sporadic and familial Alzheimer’s disease using induced pluripotent stem cells. Nature 482, 216–220 (2012). https://doi.org/10.1038/nature10821

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature10821

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