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:

PTEN recruitment controls synaptic and cognitive function in Alzheimer's models

Abstract

Dyshomeostasis of amyloid-β peptide (Aβ) is responsible for synaptic malfunctions leading to cognitive deficits ranging from mild impairment to full-blown dementia in Alzheimer's disease. Aβ appears to skew synaptic plasticity events toward depression. We found that inhibition of PTEN, a lipid phosphatase that is essential to long-term depression, rescued normal synaptic function and cognition in cellular and animal models of Alzheimer's disease. Conversely, transgenic mice that overexpressed PTEN displayed synaptic depression that mimicked and occluded Aβ-induced depression. Mechanistically, Aβ triggers a PDZ-dependent recruitment of PTEN into the postsynaptic compartment. Using a PTEN knock-in mouse lacking the PDZ motif, and a cell-permeable interfering peptide, we found that this mechanism is crucial for Aβ-induced synaptic toxicity and cognitive dysfunction. Our results provide fundamental information on the molecular mechanisms of Aβ-induced synaptic malfunction and may offer new mechanism-based therapeutic targets to counteract downstream Aβ signaling.

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: VO-OHpic semi-chronic infusion rescues the learning deficits of App/Psen1 mice.
Figure 2: PTEN activity determines the synaptic response to synthetic Aβ.
Figure 3: PTEN inhibition protects Appswe/lnd-expressing neurons.
Figure 4: Blockade of PTEN activity protects neurons from extracellular Aβ.
Figure 5: Inhibition of PTEN activity in vivo and in vitro rescues synaptic plasticity in conditions of high Aβ levels.
Figure 6: Aβ-induced local regulation of the PIP3 pathway.
Figure 7: Aβ-induced synaptic depression depends on PTEN-PDZ interaction.
Figure 8: Synthetic peptide corresponding to the PTEN-PDZ binding motif prevents Aβ-induced synaptic and cognitive deficits.

Similar content being viewed by others

References

  1. Terry, R.D. et al. Physical basis of cognitive alterations in Alzheimer's disease: synapse loss is the major correlate of cognitive impairment. Ann. Neurol. 30, 572–580 (1991).

    CAS  PubMed  Google Scholar 

  2. Selkoe, D.J. Alzheimer's disease is a synaptic failure. Science 298, 789–791 (2002).

    CAS  PubMed  Google Scholar 

  3. Jack, C.R. Jr. et al. Brain beta-amyloid measures and magnetic resonance imaging atrophy both predict time-to-progression from mild cognitive impairment to Alzheimer's disease. Brain 133, 3336–3348 (2010).

    PubMed  PubMed Central  Google Scholar 

  4. Selkoe, D.J. Resolving controversies on the path to Alzheimer's therapeutics. Nat. Med. 17, 1060–1065 (2011).

    CAS  PubMed  Google Scholar 

  5. Li, S. et al. Soluble oligomers of amyloid Beta protein facilitate hippocampal long-term depression by disrupting neuronal glutamate uptake. Neuron 62, 788–801 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Hsieh, H. et al. AMPAR removal underlies Abeta-induced synaptic depression and dendritic spine loss. Neuron 52, 831–843 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Shankar, G.M. et al. Natural oligomers of the Alzheimer amyloid-beta protein induce reversible synapse loss by modulating an NMDA-type glutamate receptor–dependent signaling pathway. J. Neurosci. 27, 2866–2875 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Shankar, G.M. et al. Amyloid-beta protein dimers isolated directly from Alzheimer's brains impair synaptic plasticity and memory. Nat. Med. 14, 837–842 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Walsh, D.M. et al. Naturally secreted oligomers of amyloid beta protein potently inhibit hippocampal long-term potentiation in vivo. Nature 416, 535–539 (2002).

    CAS  PubMed  Google Scholar 

  10. Cullen, W.K., Wu, J., Anwyl, R. & Rowan, M.J. beta-Amyloid produces a delayed NMDA receptor–dependent reduction in synaptic transmission in rat hippocampus. Neuroreport 8, 87–92 (1996).

    CAS  PubMed  Google Scholar 

  11. Lambert, M.P. et al. Diffusible, nonfibrillar ligands derived from Abeta1-42 are potent central nervous system neurotoxins. Proc. Natl. Acad. Sci. USA 95, 6448–6453 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Man, H.Y. et al. Activation of PI3-kinase is required for AMPA receptor insertion during LTP of mEPSCs in cultured hippocampal neurons. Neuron 38, 611–624 (2003).

    CAS  PubMed  Google Scholar 

  13. Peineau, S. et al. LTP inhibits LTD in the hippocampus via regulation of GSK3beta. Neuron 53, 703–717 (2007).

    CAS  PubMed  Google Scholar 

  14. Arendt, K.L. et al. PIP3 controls synaptic function by maintaining AMPA receptor clustering at the postsynaptic membrane. Nat. Neurosci. 13, 36–44 (2010).

    CAS  PubMed  Google Scholar 

  15. Jurado, S. et al. PTEN is recruited to the postsynaptic terminal for NMDA receptor-dependent long-term depression. EMBO J. 29, 2827–2840 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Holcomb, L. et al. Accelerated Alzheimer-type phenotype in transgenic mice carrying both mutant amyloid precursor protein and presenilin 1 transgenes. Nat. Med. 4, 97–100 (1998).

    CAS  PubMed  Google Scholar 

  17. Rosivatz, E. et al. A small molecule inhibitor for phosphatase and tensin homologue deleted on chromosome 10 (PTEN). ACS Chem. Biol. 1, 780–790 (2006).

    CAS  PubMed  Google Scholar 

  18. Jan, A., Hartley, D.M. & Lashuel, H.A. Preparation and characterization of toxic Abeta aggregates for structural and functional studies in Alzheimer's disease research. Nat. Protoc. 5, 1186–1209 (2010).

    CAS  PubMed  Google Scholar 

  19. Selkoe, D.J. Soluble oligomers of the amyloid beta-protein impair synaptic plasticity and behavior. Behav. Brain Res. 192, 106–113 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Welsby, P.J., Rowan, M.J. & Anwyl, R. Beta-amyloid blocks high frequency stimulation induced LTP but not nicotine enhanced LTP. Neuropharmacology 53, 188–195 (2007).

    CAS  PubMed  Google Scholar 

  21. Klyubin, I. et al. Soluble Arctic amyloid beta protein inhibits hippocampal long-term potentiation in vivo. Eur. J. Neurosci. 19, 2839–2846 (2004).

    PubMed  Google Scholar 

  22. Wang, Q., Walsh, D.M., Rowan, M.J., Selkoe, D.J. & Anwyl, R. Block of long-term potentiation by naturally secreted and synthetic amyloid beta-peptide in hippocampal slices is mediated via activation of the kinases c-Jun N-terminal kinase, cyclin-dependent kinase 5, and p38 mitogen-activated protein kinase as well as metabotropic glutamate receptor type 5. J. Neurosci. 24, 3370–3378 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Chen, Q.S., Wei, W.Z., Shimahara, T. & Xie, C.W. Alzheimer amyloid beta-peptide inhibits the late phase of long-term potentiation through calcineurin-dependent mechanisms in the hippocampal dentate gyrus. Neurobiol. Learn. Mem. 77, 354–371 (2002).

    CAS  PubMed  Google Scholar 

  24. Ortega-Molina, A. et al. Pten positively regulates brown adipose function, energy expenditure, and longevity. Cell Metab. 15, 382–394 (2012).

    CAS  PubMed  Google Scholar 

  25. Gerges, N.Z., Brown, T.C., Correia, S.S. & Esteban, J.A. Analysis of Rab protein function in neurotransmitter receptor trafficking at hippocampal synapses. Methods Enzymol. 403, 153–166 (2005).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  27. Citron, M., Teplow, D.B. & Selkoe, D.J. Generation of amyloid beta protein from its precursor is sequence specific. Neuron 14, 661–670 (1995).

    CAS  PubMed  Google Scholar 

  28. Jacobsen, J.S. et al. Early-onset behavioral and synaptic deficits in a mouse model of Alzheimer's disease. Proc. Natl. Acad. Sci. USA 103, 5161–5166 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Knafo, S. et al. Widespread changes in dendritic spines in a model of Alzheimer's disease. Cereb. Cortex 19, 586–592 (2009).

    CAS  PubMed  Google Scholar 

  30. Wei, W. et al. Amyloid beta from axons and dendrites reduces local spine number and plasticity. Nat. Neurosci. 13, 190–196 (2010).

    CAS  PubMed  Google Scholar 

  31. Spires, T.L. et al. Dendritic spine abnormalities in amyloid precursor protein transgenic mice demonstrated by gene transfer and intravital multiphoton microscopy. J. Neurosci. 25, 7278–7287 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Maehama, T. & Dixon, J.E. The tumor suppressor, PTEN/MMAC1, dephosphorylates the lipid second messenger, phosphatidylinositol 3,4,5-trisphosphate. J. Biol. Chem. 273, 13375–13378 (1998).

    CAS  PubMed  Google Scholar 

  33. He, J. et al. Proteomic analysis of beta1-adrenergic receptor interactions with PDZ scaffold proteins. J. Biol. Chem. 281, 2820–2827 (2006).

    CAS  PubMed  Google Scholar 

  34. Wu, Y. et al. Interaction of the tumor suppressor PTEN/MMAC with a PDZ domain of MAGI3, a novel membrane-associated guanylate kinase. J. Biol. Chem. 275, 21477–21485 (2000).

    CAS  PubMed  Google Scholar 

  35. Wu, X. et al. Evidence for regulation of the PTEN tumor suppressor by a membrane-localized multi-PDZ domain containing scaffold protein MAGI-2. Proc. Natl. Acad. Sci. USA 97, 4233–4238 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. von Stein, W., Ramrath, A., Grimm, A., Müller-Borg, M. & Wodarz, A. Direct association of Bazooka/PAR-3 with the lipid phosphatase PTEN reveals a link between the PAR/aPKC complex and phosphoinositide signaling. Development 132, 1675–1686 (2005).

    CAS  PubMed  Google Scholar 

  37. Cissé, M. et al. Reversing EphB2 depletion rescues cognitive functions in Alzheimer model. Nature 469, 47–52 (2011).

    PubMed  Google Scholar 

  38. Jo, J. et al. A(1-42) inhibition of LTP is mediated by a signaling pathway involving caspase-3, Akt1 and GSK-3β. Nat. Neurosci. 14, 545–547 (2011).

    CAS  PubMed  Google Scholar 

  39. Hongpaisan, J., Sun, M.K. & Alkon, D.L. PKC ɛ activation prevents synaptic loss, Aβ elevation, and cognitive deficits in Alzheimer's disease transgenic mice. J. Neurosci. 31, 630–643 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Ma, T. et al. Dysregulation of the mTOR pathway mediates impairment of synaptic plasticity in a mouse model of Alzheimer's disease. PLoS One 5, e12845 (2010).

    PubMed  PubMed Central  Google Scholar 

  41. Kwak, Y.D. et al. NO signaling and S-nitrosylation regulate PTEN inhibition in neurodegeneration. Mol. Neurodegener. 5, 49 (2010).

    PubMed  PubMed Central  Google Scholar 

  42. Talbot, K. et al. Demonstrated brain insulin resistance in Alzheimer's disease patients is associated with IGF-1 resistance, IRS-1 dysregulation, and cognitive decline. J. Clin. Invest. 122, 1316–1338 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Zhang, X. et al. Tumor-suppressor PTEN affects tau phosphorylation, aggregation, and binding to microtubules. FASEB J. 20, 1272–1274 (2006).

    CAS  PubMed  Google Scholar 

  44. Griffin, R.J. et al. Activation of Akt/PKB, increased phosphorylation of Akt substrates and loss and altered distribution of Akt and PTEN are features of Alzheimer's disease pathology. J. Neurochem. 93, 105–117 (2005).

    CAS  PubMed  Google Scholar 

  45. Pei, J.J. et al. Role of protein kinase B in Alzheimer's neurofibrillary pathology. Acta Neuropathol. 105, 381–392 (2003).

    CAS  PubMed  Google Scholar 

  46. Freude, S. et al. Neuronal IGF-1 resistance reduces Abeta accumulation and protects against premature death in a model of Alzheimer's disease. FASEB J. 23, 3315–3324 (2009).

    CAS  PubMed  Google Scholar 

  47. Cohen, E. et al. Reduced IGF-1 signaling delays age-associated proteotoxicity in mice. Cell 139, 1157–1169 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. De Felice, F.G., Lourenco, M.V. & Ferreira, S.T. How does brain insulin resistance develop in Alzheimer's disease? Alzheimers Dement. 10 (suppl. 1) S26–S32 (2014).

    PubMed  Google Scholar 

  49. Ehrengruber, M.U. et al. Recombinant Semliki Forest virus and Sindbis virus efficiently infect neurons in hippocampal slice cultures. Proc. Natl. Acad. Sci. USA 96, 7041–7046 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Torres, J. et al. Heterogeneous lack of expression of the tumour suppressor PTEN protein in human neoplastic tissues. Eur. J. Cancer 37, 114–121 (2001).

    CAS  PubMed  Google Scholar 

  51. Valiente, M. et al. Binding of PTEN to specific PDZ domains contributes to PTEN protein stability and phosphorylation by microtubule-associated serine/threonine kinases. J. Biol. Chem. 280, 28936–28943 (2005).

    CAS  PubMed  Google Scholar 

  52. Hood, C.A. et al. Fast conventional Fmoc solid-phase peptide synthesis with HCTU. J. Pept. Sci. 14, 97–101 (2008).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank F. Valdivieso (Centro de Biología Molecular “Severo Ochoa”) for the Appswe/lnd construct, W. Klein (Northwestern University) for the Aβ antibody (NU-1), D. Walsh (Harvard Institutes of Medicine) for expert advice and providing protocols, and members of the Esteban laboratory for critical reading of this manuscript. This work was supported by grants from the Spanish Ministry of Economy and Competitiveness (CSD-2010-00045, SAF-2011-24730 and SAF2014-57233-R to J.A.E.; SAF2010-15676, SAF2013-43902-R and SAF2015-62540-ERC to S.K.; SAF-2009-09129 to C.V.; SAF2009-12249-C02-01 to F.W.; CSD2010-00045 and SAF2010-14906 to C.G.D.; SAF2013-48812-R to R.P.). The laboratory of S.K. is supported by a grant from Alzheimer's Association (NIRG-13-279533), from the Basque Ministry of Health (2013111138), from the University of the Basque Country (EHUrOPE14/03) and from Ikerbasque foundation. The laboratory of F.W. is also supported by CIBERNED (an initiative of ISCIII) and by EU-FP7-2009-CT222887 grant. M.S. is funded by grants from the MINECO, European Union (ERC Advanced Grant), Regional Government of Madrid, Botín Foundation, Ramón Areces Foundation, and AXA Foundation. A.M.C. is funded by grants R01CA095063 and R21CA133669 from the US National Institutes of Health. T.W. is funded from the Federal Office for Scientific Affairs (IUAP P6/43) and Flemish Government's Methusalem Grant. N.Z.G. is funded from the US National Institute on Aging (AG032320), Alzheimer's Association and American Health Assistance Foundation (AHAF). S.K. was the recipient of a “Ramón y Cajal” contract from the Spanish Ministry of Science and Innovation and is now an IkerBasque Research Professor. C.S.-P. is a recipient of an FPI scholarship from the Spanish Ministry of Economy and Competitiveness (BES-2011-043464). J.M. is the recipient of a predoctoral fellowship (PRE_2014_1_285) from Gobierno Vasco, Departamento de Educación (Basque Country, Spain).

Author information

Authors and Affiliations

Authors

Contributions

S.K. performed and analyzed most of the in vitro experiments. C.S.-P. performed most biochemical and imaging experiments. J.E.D. performed the EGFP-GSK3β imaging experiments. T.W., E.P., I.D. and L.L.-M. performed some of the biochemical and imaging experiments under the supervision of C.G.D. I.P.-P. performed some of the behavioral experiments. E.K., E.B.F. and H.M.C. designed and synthesized the PTEN peptides under the supervision of M.R.S. The specificity of PTEN-PDZ peptide using PDZ domain overlays was carried out by L.M. under the supervision of R.A.H. GST pull-down experiments were carried out by J.M. under the supervision of R.P. N.Z.G. and K.K. performed some of the patch-clamp experiments. A.M.C., A.O.-M. and M.S. developed transgenic PTEN mice. L.O.-G., F.W. and J.V. analyzed, characterized and maintained the App/Psen1 mouse colonies. C.V. designed and supervised all in vivo manipulations. J.A.E., S.K. and C.V. designed the study and wrote the paper.

Corresponding authors

Correspondence to Shira Knafo, César Venero or José A Esteban.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 PCR-genotyping of the three mouse models used in this study and controls for behavioral experiments after semi-chronic Pten inhibition.

a-c. DNA from App/Psen1 (a), Ptentg (b) and PtenΔPDZ (c) was analyzed by PCR with the primers described in Supplemental Information. Representative PCR reactions from transgenic animals and wild-type littermates are shown for each case. d. Total time spent exploring in the novel object location task, by App/Psen1 animals and WT littermate controls, with or without VO-OHpic treatment. e. Percentage freezing displayed by App/Psen1 animals and WT littermate controls, with or without VO-OHpic treatment, before fear conditioning (white columns) or after contextual fear conditioning, before tone presentation in the memory test (see Methods). f. Representative western blot of hippocampal homogenates prepared from App/Psen1 mice and their WT littermates after semi-chronic (3-4 weeks) treatment with VO-OHpic. Uncropped version of the western blot is shown in Supplementary Fig. 10.

Supplementary Figure 2 VO-OHpic specificity, characterization of Aβ assemblies and effect on electrophysiological responses

a. Representative western blots of phospho-Akt (pAkt, Thr308), total-Akt (tAkt), phospho-Tyrosine (pTyr), and actin as loading control, from acute hippocampal slices incubated with 50 nM VO-OHpic for 30, 60 or 120 min. Uncropped versions of the western blots are shown in Supplementary Fig. 10. b. Quantification of the experiment described in (a). Relative levels (normalized to vehicle) of pAkt, tAkt, pTyr and actin after treatment with VO-OHpic for different periods of time, as indicated. c. Kinetics of Aβ aggregation resulting from incubation of synthetic Aβ42 alone or with either of the PTEN inhibitors used in this study (VO-OHpic and bPV(HOpic)) as measured with the Thioflavin T assay. The mean fluorescence (at 490nm) ± SEM (in arbitrary units) is plotted against the incubation period (at 37 °C). Each point represents the average of 3 independent experiments d. Aβ42 peptide (final concentration 258 μM) was prepared as described in Methods and incubated for different periods of time at room temperature. The lane of 5 min (5’) of incubation represents the Aβ42 assemblies used for our electrophysiological and imaging experiments. Bands were visualized by western blot analysis, probed with the N-terminal anti-Aβ antibody, 6E10, to identify SDS-stable Aβ42 species. e. Representative electron micrographs of negatively stained Aβ42 showing time-dependent assembly formation (see Methods, protocol I). Note the heterogeneous mixtures of aggregates of different sizes and morphologies and the predominant fibril formation at long incubation times (52 h). f. Representative electron micrographs of negatively stained Aβ42 assemblies, 5 min after preparation (see Methods, protocol II). g. Top: Input-output curves of field excitatory postsynaptic potentials (fEPSPs) plotted against the amplitude of the fiber volley, evoked by stimulation of Schaffer Collaterals, from slices treated with vehicle or Aβ42 assemblies (2 h incubation), as indicated. Bottom: The same data showing only low values of fiber volley. h. Paired-pulse facilitation (PPF) in Aβ- and vehicle-treated hippocampal slices. The values denote the ratio of the second fEPSP amplitude to the first fEPSP amplitude. PPF was tested for 10-, 20-, 50-, 100-, and 400-ms interstimulus intervals. Insets. Sample trace of fEPSP with an interstimulus interval of 50 ms. N, number of slices/mice. Scale bars: 0.1 mV, 50 ms. i. Evaluation of TrkB signaling (monitored by TrkB phosphorylation) from slices overexpressing GFP or Appswe/lnd, with or without PTEN inhibition by VO-OHpic incubation, as indicated.

Supplementary Figure 3 Scatter plot analyses for the effects of App expression on synaptic transmission in organotypic hippocampal slices

a. Percentage of neurons overexpressing Appswe/lnd with visible intracellular Aβ. b. Scatter plots of AMPAR- and NMDAR-mediated EPSCs recorded from pairs of neurons infected with Appswe/lnd–EGFP and neighboring uninfected neurons. Each pair of infected-uninfected neuron is represented with a single point. Black circles represent the mean values of EPSCs amplitudes. c. Representative confocal projection image of neurons co-expressing AppMV and EGFP. d. Example traces from uninfected and AppMV-EGFP expressing neurons recorded at -60 mV (AMPAR EPSCs), and +40 mV (NMDAR EPSCs). e. Scatter plots of AMPAR- and NMDAR-mediated EPSCs recorded from pairs of neurons infected with AppMV–EGFP and neighboring uninfected neurons. Each pair of infected-uninfected neuron is represented with a single point. f. Scatter plots of AMPAR- and NMDAR-mediated EPSCs recorded from pairs of neurons infected with Appswe/lnd–EGFP and neighboring uninfected neurons all treated with PTEN inhibitors (15 nM bPV(HOpic) –dark grey circles- or 50 nM VO-OHpic –light grey circles-). Each pair of infected-uninfected neuron is represented with a single point. Black circles represent the mean values of EPSCs amplitudes.

Supplementary Figure 4 Electrophysiology controls and Aβ assemblies

a, c. Time course of AMPAR-mediated synaptic responses of the control (non-induced) pathway from LTP experiments shown in Fig. 4g-j, without (a) and with (c) PTEN inhibition (50 nM VO-OHpic, 5 h after virus injection and during recordings), from neurons in control (uninjected) slices, Appswe/lnd-infected neurons or neighboring uninfected neurons, as indicated. b. Time course of AMPAR-mediated synaptic responses before and after LTP induction from GFP-infected neurons or neighboring uninfected neurons. d. Biochemical characterization of Aβ42 assembly formation by western blot analysis (left panel) and electron microscopy (right panel). Aβ42 was prepared from lyophilized peptide (Invitrogen, #03-112), dissolved in water at 6 mg/ml, and then diluted to 1 mg/ml with PBS and incubated at 37ºC for 36 h (see Methods, protocol II). e. Representative confocal projection image of neurons in organotypic hippocampal slices infected with PTEN-C124S–EGFP (blue channel, fluorescence excluded from the nucleus) or Appswe/lnd-IRES-EGFP (blue channel, fluorescence in the nucleus). To visualize Aβ production, slices were fixed, permeabilized and immunostained for Aβ (NU-1 antibody, green channel). Note that neurons expressing Appswe/lnd (blue channel, fluorescence in the nucleus) are positive for Aβ staining (green signal), while neurons expressing PTEN-C124S (blue channel, fluorescence excluded from the nucleus, white arrowheads) are negative for Aβ staining. See Fig. 5d for a schematic representation of the experimental configuration. f. Scatter plots of AMPAR-mediated EPSCs recorded from pairs of neurons infected with PTEN-C124S–EGFP and neighboring uninfected neurons, all exposed to extracellular Aβ secreted from neighboring Appswe/lnd-expressing neurons (gray circles) or added exogenously (white circles). Each pair of infected-uninfected neuron is represented with a single point. g. Time course of AMPAR-mediated synaptic responses of the control pathway from LTP experiments shown in Fig. 5f, g, with or without Aβ exposure. h. Absolute current flow (holding current) during LTP experiments carried out in whole-cell under voltage-clamp configuration. These data correspond to experiments shown in Fig. 4g-j and Fig. 5f, g of the main text.

Supplementary Figure 5 PTEN recruitment to synaptic compartments

a. Confocal projection image of a primary hippocampal neuron (21 days in vitro) expressing EGFP-tagged WT (full-length) PTEN before (top) and 1 h after treatment with 4 μM synthetic Aβ42 assemblies (bottom). See Methods for preparation of primary neuronal cultures and imaging techniques. b. Quantification of time lapse imaging of the spine/dendrite ratio of WT EGFP-PTEN up-to 60 min after 1 μM Aβ42 application. N represents number of spines from 3 cells analyzed on 3 independent experiments. c. Top: Representative blot of PTEN amount on synaptosomal preparation prepared from hippocampal slices treated with Aβ for different periods of time. Uncropped versions of the western blots are shown in Supplementary Fig. 10. Bottom: Quantification of the blots represented at the top panel. N=5 rats.

Supplementary Figure 6 Aβ overproduction does not globally alter the PIP3 pathway

a. Representative blot of PTEN (left) and quantification of PTEN levels (right) from extracts of hippocampal slice cultures injected with Appswe/lnd–EGFP Sindbis virus and uninfected slices. b, c. Representative blot (left) and quantification (right) of phosphorylated and total AKT (b: pAKT(T308), tAKT), phosphorylated and total GSK3β (c: pGSK3β(S9), tGSK3β), and tubulin from extracts of hippocampal slice cultures injected with Appswe/lnd-EGFP Sindbis virus or control slices. d-f. Similar to a-c with extracts of hippocampi from App/Psen1 mice and their WT littermates. N represents the number of experiments. Statistical significance was calculated according to the Mann-Whitney test. g. Top: High magnification images of dendritic spines from neurons expressing WT EGFP-GSK3β. Bottom: Quantification of time lapse imaging of the spine/dendrite ratio of WT EGFP-GSK3β up-to 60 min after Aβ42 application. N represents number of spines from 3 cells analyzed on 3 independent experiments. h. Top: Representative blot of pAKT and tAKT amount on synaptosomal preparation prepared from hippocampal slices treated with Aβ for different periods of time. Bottom: Quantification of the blots represented at the top panel. N=6 rats. Uncropped versions of the western blots in panels a-f and h are shown in Supplementary Fig. 10.

Supplementary Figure 7 NMDAR-dependent LTD in WT and PtenΔPDZ mice

a, b. LTD was induced in WT acute hippocampal slices (900 pulses at 1 Hz) in the absence (a) or in the presence of a saturating concentration (50 μM) of AP5 (b). Some slices were incubated in 1 μM Aβ assemblies, as indicated. c. Similar to A, with acute slices from PtenΔPDZ mice. d. Quantification of the average fEPSP maximal slope, at 50-60 minutes after LTD induction, from the experiments shown in a-c. P value was determined with Wilcoxon test relative to baseline.

Supplementary Figure 8 Chemical structure and biochemical analysis of PTEN synthetic peptides and their permeability in neurons

a. Top panels: Chemical structure and Liquid Chromatography-Mass Spectrometry (LC-MS) analysis for each of the peptides used for this study. Bottom panels: Reversed-Phase High-Performance Liquid Chromatography (RP-HPLC) analysis of each peptide. b. Primary hippocampal neurons (21 days in vitro) were incubated with Fluorescein-tagged PTEN-PDZ peptide (10 μM) dissolved in DMSO (final concentration 0.1%). Top: Confocal projection image of an individual neuron before (0’) and after (2’and 50’) the addition of PTEN-PDZ peptide, demonstrating initial accumulation of the peptide at the plasma membrane (2’), followed by diffusion into the cytosol (50’). Bottom: brightfield images of the same neuron. c. N-terminus biotinylated PTEN PDZ peptide was overlaid onto an array of PDZ domains. Bound peptide was visualized with the ABC kit from Vectasatin, based on its biotin modification. “Control” represents signal detected without peptide incubation. Uncropped version of the blot is shown in Supplementary Fig. 10. d. GST pull-down of recombinant PTEN bound to a GST fusion of the PDZ2 domain of hDlg, or to GST alone (left-most lane). GST beads were incubated with recombinant PTEN in the presence of 200 μM of the indicated peptide, or in the absence of peptide, as control. Pulled-down PTEN was then visualized by western blot, with an anti-PTEN antibody. Binding is normalized to the conditions without peptide incubation. Uncropped version of the western blot is shown in Supplementary Fig. 10. e. Top: superimposed representative fEPSPs evoked by stimulation of Schaffer Collaterals after 2 h incubation in Aβ42 assemblies (or vehicle control) with or without preincubation (0.5 h) with 10 μM scrambled peptide at 10, 50, 100, 150 and 200 μA of stimulation intensity. Bottom: Input-output curves of fEPSPs. P values were determined with two-way ANOVA with Bonferroni post-tests (results without peptide preincubation are replotted from Fig. 8e as lines). Scale bars: 0.5 mV, 20 ms. Data are represented as mean +/− SEM.

Supplementary Figure 9 Uncropped versions of blots from main figures

Uncropped versions of western blots from Figure panels 1d, 1e, 2d, 3b, 3c, 3e and 7b are shown, as indicated.

Supplementary Figure 10 Uncropped versions of blots from supplementary figures

Uncropped versions of western blots from Supplementary Figure panels 1f, 2a, 5c, 6a-f, 6h, 8d, and overlay blot from Supplementary Figure 8c are shown, as indicated.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Knafo, S., Sánchez-Puelles, C., Palomer, E. et al. PTEN recruitment controls synaptic and cognitive function in Alzheimer's models. Nat Neurosci 19, 443–453 (2016). https://doi.org/10.1038/nn.4225

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nn.4225

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