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:

Gain of toxic apolipoprotein E4 effects in human iPSC-derived neurons is ameliorated by a small-molecule structure corrector

Abstract

Efforts to develop drugs for Alzheimer's disease (AD) have shown promise in animal studies, only to fail in human trials, suggesting a pressing need to study AD in human model systems. Using human neurons derived from induced pluripotent stem cells that expressed apolipoprotein E4 (ApoE4), a variant of the APOE gene product and the major genetic risk factor for AD, we demonstrated that ApoE4-expressing neurons had higher levels of tau phosphorylation, unrelated to their increased production of amyloid-β (Aβ) peptides, and that they displayed GABAergic neuron degeneration. ApoE4 increased Aβ production in human, but not in mouse, neurons. Converting ApoE4 to ApoE3 by gene editing rescued these phenotypes, indicating the specific effects of ApoE4. Neurons that lacked APOE behaved similarly to those expressing ApoE3, and the introduction of ApoE4 expression recapitulated the pathological phenotypes, suggesting a gain of toxic effects from ApoE4. Treatment of ApoE4-expressing neurons with a small-molecule structure corrector ameliorated the detrimental effects, thus showing that correcting the pathogenic conformation of ApoE4 is a viable therapeutic approach for ApoE4-related AD.

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

Fig. 1: Human ApoE4/4 neurons generate more APOE fragments, have higher p-tau levels and produce more Aβ than human ApoE3/3 neurons.
Fig. 2: Increased p-tau levels in ApoE4/4 neurons are independent of higher Aβ production.
Fig. 3: ApoE4 causes GABAergic neuron degeneration and/or loss in hiPSC-derived neuronal cultures.
Fig. 4: AD-related pathologies in human ApoE4/4 neurons are specifically induced by ApoE4.
Fig. 5: ApoE4 confers a gain of toxic effects in hiPSC-derived neurons.
Fig. 6: The gain of toxic effects of ApoE4 in hiPSC-derived neurons can be ameliorated by a small-molecule structure corrector.

Similar content being viewed by others

References

  1. Huang, Y. & Mucke, L. Alzheimer mechanisms and therapeutic strategies. Cell 148, 1204–1222 (2012).

    Article  CAS  Google Scholar 

  2. Golde, T. E., Schneider, L. S. & Koo, E. H. Anti-Aβ therapeutics in Alzheimer's disease: the need for a paradigm shift. Neuron 69, 203–213 (2011).

    Article  CAS  Google Scholar 

  3. Corder, E. H. et al. Gene dose of apolipoprotein E type 4 allele and the risk of Alzheimer's disease in late-onset families. Science 261, 921–923 (1993).

    Article  CAS  Google Scholar 

  4. Saunders, A. M. et al. Association of apolipoprotein E allele ε4 with late-onset familial and sporadic Alzheimer's disease. Neurology 43, 1467–1472 (1993).

    Article  CAS  Google Scholar 

  5. Farrer, L. A. et al. APOE and Alzheimer Disease Meta Analysis Consortium. Effects of age, sex and ethnicity on the association between apolipoprotein E genotype and Alzheimer's disease. A meta-analysis. J. Am. Med. Assoc. 278, 1349–1356 (1997).

    Article  CAS  Google Scholar 

  6. Genin, E. et al. APOE and Alzheimer's disease: a major gene with semi-dominant inheritance. Mol. Psychiatry 16, 903–907 (2011).

    Article  CAS  Google Scholar 

  7. Huang, Y. Aβ-independent roles of apolipoprotein E4 in the pathogenesis of Alzheimer's disease. Trends Mol. Med. 16, 287–294 (2010).

    Article  CAS  Google Scholar 

  8. Mahley, R. W. & Huang, Y. Apolipoprotein E sets the stage: response to injury triggers neuropathology. Neuron 76, 871–885 (2012).

    Article  CAS  Google Scholar 

  9. Ashe, K. H. & Zahs, K. R. Probing the biology of Alzheimer's disease in mice. Neuron 66, 631–645 (2010).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  11. Yahata, N. et al. Anti-Aβ drug screening platform using human iPS-cell-derived neurons for the treatment of Alzheimer's disease. PLoS One 6, e25788 (2011).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  13. Almeida, S. et al. Induced pluripotent stem cell models of progranulin-deficient frontotemporal dementia uncover specific reversible neuronal defects. Cell Rep. 2, 789–798 (2012).

    Article  CAS  Google Scholar 

  14. 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  Google Scholar 

  15. Liu, Q. et al. Effect of potent γ-secretase modulator in human neurons derived from multiple presenilin-1 induced pluripotent stem cell mutant carriers. JAMA Neurol. 71, 1481–1489 (2014).

    Article  Google Scholar 

  16. Takahashi, K. & Yamanaka, S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 126, 663–676 (2006).

    Article  CAS  Google Scholar 

  17. 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 

  18. Brecht, W. J. et al. Neuron-specific apolipoprotein E4 proteolysis is associated with increased tau phosphorylation in brains of transgenic mice. J. Neurosci. 24, 2527–2534 (2004).

    Article  CAS  Google Scholar 

  19. Harris, F. M. et al. Carboxyl-terminal-truncated apolipoprotein E4 causes Alzheimer's-disease-like neurodegeneration and behavioral deficits in transgenic mice. Proc. Natl Acad. Sci. USA 100, 10966–10971 (2003).

    Article  CAS  Google Scholar 

  20. Hoover, B. R. et al. Tau mislocalization to dendritic spines mediates synaptic dysfunction independently of neurodegeneration. Neuron 68, 1067–1081 (2010).

    Article  CAS  Google Scholar 

  21. Thies, E. & Mandelkow, E. M. Missorting of tau in neurons causes degeneration of synapses that can be rescued by the kinase MARK2 (Par-1). J. Neurosci. 27, 2896–2907 (2007).

    Article  CAS  Google Scholar 

  22. Woodruff, G. et al. The presenilin-1 ΔE9 mutation results in reduced γ-secretase activity, but not total loss of PS1 function, in isogenic human stem cells. Cell Rep. 5, 974–985 (2013).

    Article  CAS  Google Scholar 

  23. Castellano, J. M. et al. Human APOE isoforms differentially regulate brain amyloid-β peptide clearance. Sci. Transl. Med. 3, 89ra57 (2011).

    Article  CAS  Google Scholar 

  24. 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 

  25. Jin, M. et al. Soluble amyloid β-protein dimers isolated from Alzheimer cortex directly induce Tau hyperphosphorylation and neuritic degeneration. Proc. Natl. Acad. Sci. USA 108, 5819–5824 (2011).

    Article  CAS  Google Scholar 

  26. 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 

  27. Nicholas, C. R. et al. Functional maturation of hPSC-derived forebrain interneurons requires an extended timeline and mimics human neural development. Cell Stem Cell 12, 573–586 (2013).

    Article  CAS  Google Scholar 

  28. Ahn, S., Kim, T. G., Kim, K. S. & Chung, S. Differentiation of human pluripotent stem cells into medial ganglionic eminence versus caudal ganglionic eminence cells. Methods 101, 103–112 (2016).

    Article  CAS  Google Scholar 

  29. Fong, H. et al. Genetic correction of tauopathy phenotypes in neurons derived from human induced pluripotent stem cells. Stem Cell Rep. 1, 226–234 (2013).

    Article  CAS  Google Scholar 

  30. Mak, A. C. et al. Effects of the absence of apolipoprotein E on lipoproteins, neurocognitive function and retinal function. JAMA Neurol. 71, 1228–1236 (2014).

    Article  Google Scholar 

  31. Xu, Q. et al. Profile and regulation of apolipoprotein E (APOE) expression in the CNS in mice with targeting of green fluorescent protein gene to the Apoe locus. J. Neurosci. 26, 4985–4994 (2006).

    Article  CAS  Google Scholar 

  32. Xu, Q. et al. Intron-3 retention and splicing controls neuronal expression of apolipoprotein E in the CNS. J. Neurosci. 28, 1452–1459 (2008).

    Article  CAS  Google Scholar 

  33. Xu, P.-T. et al. Specific regional transcription of apolipoprotein E in human brain neurons. Am. J. Pathol. 154, 601–611 (1999).

    Article  CAS  Google Scholar 

  34. Ulrich, J. D. et al. In vivo measurement of apolipoprotein E from the brain interstitial fluid using microdialysis. Mol. Neurodegener. 8, 13 (2013).

    Article  CAS  Google Scholar 

  35. Mahley, R. W., Weisgraber, K. H. & Huang, Y. Apolipoprotein E4: a causative factor and therapeutic target in neuropathology, including Alzheimer's disease. Proc. Natl. Acad. Sci. USA 103, 5644–5651 (2006).

    Article  CAS  Google Scholar 

  36. Chen, H. K. et al. Small-molecule structure correctors abolish detrimental effects of apolipoprotein E4 in cultured neurons. J. Biol. Chem. 287, 5253–5266 (2012).

    Article  CAS  Google Scholar 

  37. Brodbeck, J. et al. Structure-dependent impairment of intracellular apolipoprotein E4 trafficking and its detrimental effects are rescued by small-molecule structure correctors. J. Biol. Chem. 286, 17217–17226 (2011).

    Article  CAS  Google Scholar 

  38. Mahley, R. W. & Huang, Y. Small-molecule structure correctors target abnormal protein structure and function: structure corrector rescue of apolipoprotein-E4-associated neuropathology. J. Med. Chem. 55, 8997–9008 (2012).

    Article  CAS  Google Scholar 

  39. Bien-Ly, N., Gillespie, A. K., Walker, D., Yoon, S. Y. & Huang, Y. Reducing human apolipoprotein E levels attenuates age-dependent Aβ accumulation in mutant human amyloid precursor protein transgenic mice. J. Neurosci. 32, 4803–4811 (2012).

    Article  CAS  Google Scholar 

  40. Kim, J., Basak, J. M. & Holtzman, D. M. The role of apolipoprotein E in Alzheimer's disease. Neuron 63, 287–303 (2009).

    Article  CAS  Google Scholar 

  41. Huynh, T. V. et al. Age-dependent effects of apoE reduction using antisense oligonucleotides in a model of β-amyloidosis. Neuron 96, 1013–1023.e4 (2017).

    Article  CAS  Google Scholar 

  42. Liu, C. C. et al. ApoE4 accelerates early seeding of amyloid pathology. Neuron 96, 1024–1032.e3 (2017).

    Article  CAS  Google Scholar 

  43. Huang, Y. A., Zhou, B., Wernig, M. & Südhof, T. C. ApoE2, ApoE3 and ApoE4 differentially stimulate APP transcription and Aβ secretion. Cell 168, 427–441.e21 (2017).

    Article  CAS  Google Scholar 

  44. Kamenetz, F. et al. APP processing and synaptic function. Neuron 37, 925–937 (2003).

    Article  CAS  Google Scholar 

  45. Cirrito, J. R. et al. Synaptic activity regulates interstitial fluid amyloid-β levels in vivo. Neuron 48, 913–922 (2005).

    Article  CAS  Google Scholar 

  46. LaDu, M. J. et al. Self-assembly of HEK-cell-secreted ApoE particles resembles ApoE enrichment of lipoproteins as a ligand for the LDL-receptor-related protein. Biochemistry 45, 381–390 (2006).

    Article  CAS  Google Scholar 

  47. Shi, Y. et al. ApoE4 markedly exacerbates Tau-mediated neurodegeneration in a mouse model of tauopathy. Nature 549, 523–527 (2017).

    Article  Google Scholar 

  48. Andrews-Zwilling, Y. et al. Apolipoprotein E4 causes age- and Tau-dependent impairment of GABAergic interneurons, leading to learning and memory deficits in mice. J. Neurosci. 30, 13707–13717 (2010).

    Article  CAS  Google Scholar 

  49. Leung, L. et al. Apolipoprotein E4 causes age- and sex-dependent impairments of hilar GABAergic interneurons and learning and memory deficits in mice. PLoS One 7, e53569 (2012).

    Article  CAS  Google Scholar 

  50. Li, G. et al. GABAergic interneuron dysfunction impairs hippocampal neurogenesis in adult apolipoprotein E4 knock-in mice. Cell Stem Cell 5, 634–645 (2009).

    Article  CAS  Google Scholar 

  51. Tong, L. M. et al. Inhibitory interneuron progenitor transplantation restores normal learning and memory in ApoE4 knock-in mice without or with Aβ accumulation. J. Neurosci. 34, 9506–9515 (2014).

    Article  Google Scholar 

  52. Nuriel, T. et al. Neuronal hyperactivity due to loss of inhibitory tone in APOE4 mice lacking Alzheimer's-disease-like pathology. Nat. Commun. 8, 1464 (2017).

    Article  Google Scholar 

  53. Knoferle, J. et al. Apolipoprotein E4 produced in GABAergic interneurons causes learning and memory deficits in mice. J. Neurosci. 34, 14069–14078 (2014).

    Article  Google Scholar 

  54. Gillespie, A. K. et al. Apolipoprotein E4 causes age-dependent disruption of slow γ-oscillations during hippocampal sharp-wave ripples. Neuron 90, 740–751 (2016).

    Article  CAS  Google Scholar 

  55. Liu, C. C., Liu, C. C., Kanekiyo, T., Xu, H. & Bu, G. Apolipoprotein E and Alzheimer's disease: risk, mechanisms and therapy. Nat. Rev. Neurol. 9, 106–118 (2013).

    Article  CAS  Google Scholar 

  56. Takahashi, K., Okita, K., Nakagawa, M. & Yamanaka, S. Induction of pluripotent stem cells from fibroblast cultures. Nat. Protoc. 2, 3081–3089 (2007).

    Article  CAS  Google Scholar 

  57. Chambers, S. M. et al. Highly efficient neural conversion of human ES and iPS cells by dual inhibition of SMAD signaling. Nat. Biotechnol. 27, 275–280 (2009).

    Article  CAS  Google Scholar 

  58. Hu, B. Y. & Zhang, S. C. Differentiation of spinal motor neurons from pluripotent human stem cells. Nat. Protoc. 4, 1295–1304 (2009).

    Article  CAS  Google Scholar 

  59. Krencik, R., Weick, J. P., Liu, Y., Zhang, Z. J. & Zhang, S. C. Specification of transplantable astroglial subtypes from human pluripotent stem cells. Nat. Biotechnol. 29, 528–534 (2011).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank A. Karydas for assistance with skin biopsies, S. Ordway and T. Pak for editorial assistance and P. Davies (Albert Einstein College of Medicine) for the PHF1 antibody. This work was supported by funding from the US National Institutes of Health (grant nos. AG048030 (Y.H.), AG048017 (Y.H.), AG056305 (Y.H.) and AG023501 (B.L.M. and Y.H.)) and the California Institute for Regenerative Medicine (grant nos. RN2-00952 and TRAN1-09394; both to Y.H.) and by a gift from the Roddenberry Foundation. C.W. was partially supported by a fellowship from the California Institute for Regenerative Medicine.

Author information

Authors and Affiliations

Authors

Contributions

C.W. and Y.H. designed and coordinated the study; C.W. performed most of the studies and data analyses; R.N. performed all MGE studies and related data analysis; Q.X. and D.J. helped with off-target analysis of gene editing and work with APOE-expressing lentiviral vectors and performed some western blots; D.W. designed and prepared the APOE-ε3 donor DNA for gene editing; D.W. and S.Y.Y. helped with genetic screening; M.E.B. helped with the cell culture; H.Y. helped with MGE studies and image collection; G.L. helped with miPSC studies; Z.A.M., B.L.M. and M.J.M. provided the APOE-null human skin biopsy; and C.W. and Y.H. wrote the manuscript.

Corresponding author

Correspondence to Yadong Huang.

Ethics declarations

Competing interests

Y.H. is a cofounder and scientific advisory board member of E-Scape Bio, Inc.

Additional information

Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wang, C., Najm, R., Xu, Q. et al. Gain of toxic apolipoprotein E4 effects in human iPSC-derived neurons is ameliorated by a small-molecule structure corrector. Nat Med 24, 647–657 (2018). https://doi.org/10.1038/s41591-018-0004-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41591-018-0004-z

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