Inhibition of mTOR by rapamycin abolishes cognitive deficits and reduces amyloid-beta levels in a mouse model of Alzheimer's disease

Patricia Spilman, Natalia Podlutskaya, Matthew J Hart, Jayanta Debnath, Olivia Gorostiza, Dale Bredesen, Arlan Richardson, Randy Strong, Veronica Galvan, Patricia Spilman, Natalia Podlutskaya, Matthew J Hart, Jayanta Debnath, Olivia Gorostiza, Dale Bredesen, Arlan Richardson, Randy Strong, Veronica Galvan

Abstract

Background: Reduced TOR signaling has been shown to significantly increase lifespan in a variety of organisms [1], [2], [3], [4]. It was recently demonstrated that long-term treatment with rapamycin, an inhibitor of the mTOR pathway[5], or ablation of the mTOR target p70S6K[6] extends lifespan in mice, possibly by delaying aging. Whether inhibition of the mTOR pathway would delay or prevent age-associated disease such as AD remained to be determined.

Methodology/principal findings: We used rapamycin administration and behavioral tools in a mouse model of AD as well as standard biochemical and immunohistochemical measures in brain tissue to provide answers for this question. Here we show that long-term inhibition of mTOR by rapamycin prevented AD-like cognitive deficits and lowered levels of Abeta(42), a major toxic species in AD[7], in the PDAPP transgenic mouse model. These data indicate that inhibition of the mTOR pathway can reduce Abeta(42) levels in vivo and block or delay AD in mice. As expected from the inhibition of mTOR, autophagy was increased in neurons of rapamycin-treated transgenic, but not in non-transgenic, PDAPP mice, suggesting that the reduction in Abeta and the improvement in cognitive function are due in part to increased autophagy, possibly as a response to high levels of Abeta.

Conclusions/significance: Our data suggest that inhibition of mTOR by rapamycin, an intervention that extends lifespan in mice, can slow or block AD progression in a transgenic mouse model of the disease. Rapamycin, already used in clinical settings, may be a potentially effective therapeutic agent for the treatment of AD.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. Rapamycin abrogates memory deficits in…
Figure 1. Rapamycin abrogates memory deficits in PDAPP hAPP(J20) mice.
a, Rapamycin improves learning in PDAPP mice. While learning in both transgenic groups was impaired with respect to wild-type littermates' [**, P<0.001 for both comparisons, Bonferroni's post hoc test applied to a significant effect of genotype and treatment, F(3,120) = 29.46, P<0.0001, repeated measures two-way ANOVA], performance of rapamycin-fed PDAPP mice was improved with respect to the control-fed transgenic group only in the last day of training (#P = 0.036 for the comparison of performance between transgenic groups, Student's t test), indicating improved learning of rapamycin-fed PDAPP mice at day 4. No significant interaction was observed between day number and genotype (P = 0.96), indicating that genotype had roughly the same effect at all times during training. Although no significant interaction was observed between day number and treatment for control-treated animals (P = 0.91), a significant interaction was observed between day number and treatment for rapamycin-treated groups. The effect of rapamycin treatment became more pronounced as training progressed, as indicated by the slopes for the learning curves (m = −5.14 for rapamycin-treated as compared to m = −3.58 for control-treated PDAPP transgenic mice; m = −4 for rapamycin-treated as compared to m = −2.95 for control-treated non-transgenic mice). A trend to improved learning was observed in rapamycin-treated non-Tg mice, but this difference was not significant. Overall learning was effective in all groups [F(3,120) = 10.29, P<0.0001, repeated measures two-way ANOVA]. Inset, learning was effective in all experimental groups during cued training. b, Rapamycin restores spatial memory in PDAPP mice. While retention in control-fed PDAPP mice was impaired with respect to all other groups, as previously described, , , [P values are indicated, Tukey's multiple comparisons test applied to a significant effect of genotype (P<0.0001) in one-way ANOVA], memory in rapamycin-fed PDAPP mice was indistinguishable from that of control- or rapamycin-fed non-Tg groups. A trend to improved retention was observed in rapamycin-treated non-Tg mice, but this difference did not reach statistical significance. c and d, Rapamycin treatment does not affect non-cognitive components of behavior.c, Although transgenic groups spent more time engaged in thigmotactic swim, as described (** P<0.001, Bonferroni's post hoc test applied to a significant effect of genotype [F(3,440) = 15.04, P<0.0001, two-way ANOVA], no significant difference in percent time spent in thigmotactic swim was observed between transgenic groups. d, No significant difference in floating was observed between groups. Data are mean ± SEM.
Figure 2. Rapamycin inhibits mTOR and decreases…
Figure 2. Rapamycin inhibits mTOR and decreases Aβ42 levels in brains of PDAPP mice.
a, b and f, representative immunoblots of whole brain lysates from control- and rapamycin-treated PDAPP transgenic and non-transgenic littermate mice; c, g–k, quantitative analyses of protein or phosphoprotein levels. a–c, Levels of phosphorylated (activated) p70 were decreased in brains of rapamycin-treated non-transgenic (a) and transgenic PDAPP (b) mice (c, **, P = 0.006 and *, P = 0.01 respectively). d, rapamycin did not alter Aβ40 levels but significantly decreased soluble Aβ42 levels in the brains of transgenic PDAPP mice *, P = 0.02. Homogenates were measured at 100 mg brain tissue/ml. e, rapamycin did not alter levels of endogenous mouse Aβ40 levels in brains of non-transgenic mice. Aβ42 levels were below the detection limit of the ELISA (not shown). f, representative immunoblots of PDAPP mouse brain extracts. g–k, Quantitative analyses of APP, C99 and C83, NEP and IDE immunoreactivity in lysates of brains from control- and rapamycin-treated PDAPP mice. Data were normalized to β-actin levels. Student's t test was used to determine significance of differences between means. Data are means ± SEM.
Figure 3. Rapamycin increases autophagy in brains…
Figure 3. Rapamycin increases autophagy in brains of PDAPP mice.
a, f and h, representative immunoblots of hippocampal lysates from control- and rapamycin-treated transgenic PDAPP mice and non-transgenic littermate controls. b, g and i, quantitative analyses. a and b, LC3-II levels are decreased in hippocampi of rapamycin-treated transgenic PDAPP mice (*, P = 0.0009), but not in hippocampi of rapamycin-treated non-transgenic littermates. c and d, representative epifluorescent (c, 200×) and higher-magnification confocal (d, 600×) images of hippocampal CA1 (e, green box, region of epifluorescent images; blue box, region of confocal images) in control- and rapamycin-fed transgenic PDAPP mice stained with an anti-LC3 antibody. An increase in LC3-immunoreactive puncta was observed in CA1 projections of transgenic PDAPP mice following rapamycin administration. f and g, levels of the autophagic substrate p62SQSTM are decreased (*, P = 0.0015) in hippocampi of rapamycin-treated PDAPP transgenic mice. f, representative Western blots; g, quantitative analyses of p62SQSTM levels. h and i, Levels of phosphorylated (activated) p70 were decreased in brains of rapamycin-treated PDAPP and non-transgenic mice (*, P = 0.001 and P = 0.04 respectively). Significance of differences between group means were determined using two-tailed unpaired Student's t test. Data are means ± SEM.

References

    1. Kaeberlein M, Powers RW, 3rd, Steffen KK, Westman EA, Hu D, et al. Regulation of yeast replicative life span by TOR and Sch9 in response to nutrients. Science. 2005;310:1193–1196.
    1. Powers RW, 3rd, Kaeberlein M, Caldwell SD, Kennedy BK, Fields S. Extension of chronological life span in yeast by decreased TOR pathway signaling. Genes Dev. 2006;20:174–184.
    1. Jia K, Chen D, Riddle DL. The TOR pathway interacts with the insulin signaling pathway to regulate C. elegans larval development, metabolism and life span. Development. 2004;131:3897–3906.
    1. Vellai T, Takacs-Vellai K, Zhang Y, Kovacs AL, Orosz L, et al. Genetics: influence of TOR kinase on lifespan in C. elegans. Nature. 2003;426:620.
    1. Harrison DE, Strong R, Sharp ZD, Nelson JF, Astle CM, et al. Rapamycin fed late in life extends lifespan in genetically heterogeneous mice. Nature. 2009;460:392–395.
    1. Selman C, Tullet JM, Wieser D, Irvine E, Lingard SJ, et al. Ribosomal protein S6 kinase 1 signaling regulates mammalian life span. Science. 2009;326:140–144.
    1. McGowan E, Pickford F, Kim J, Onstead L, Eriksen J, et al. Abeta42 is essential for parenchymal and vascular amyloid deposition in mice. Neuron. 2005;47:191–199.
    1. Selkoe DJ. Alzheimer's disease is a synaptic failure. Science. 2002;298:789–791.
    1. Hsia AY, Masliah E, McConlogue L, Yu GQ, Tatsuno G, et al. Plaque-independent disruption of neural circuits in Alzheimer's disease mouse models. Proc Natl Acad Sci U S A. 1999;96:3228–3233.
    1. Mucke L, Masliah E, Yu GQ, Mallory M, Rockenstein EM, et al. High-level neuronal expression of abeta 1-42 in wild-type human amyloid protein precursor transgenic mice: synaptotoxicity without plaque formation. J Neurosci. 2000;20:4050–4058.
    1. Galvan V, Gorostiza OF, Banwait S, Ataie M, Logvinova AV, et al. Reversal of Alzheimer's-like pathology and behavior in human APP transgenic mice by mutation of Asp664. Proc Natl Acad Sci U S A. 2006;103:7130–7135.
    1. Sarbassov DD, Ali SM, Sabatini DM. Growing roles for the mTOR pathway. Curr Opin Cell Biol. 2005;17:596–603.
    1. Guertin DA, Stevens DM, Thoreen CC, Burds AA, Kalaany NY, et al. Ablation in mice of the mTORC components raptor, rictor, or mLST8 reveals that mTORC2 is required for signaling to Akt-FOXO and PKCalpha, but not S6K1. Dev Cell. 2006;11:859–871.
    1. Mizushima N. The role of the Atg1/ULK1 complex in autophagy regulation. Curr Opin Cell Biol. 2009;22:1–8.
    1. Zeng Z, Sarbassov dos D, Samudio IJ, Yee KW, Munsell MF, et al. Rapamycin derivatives reduce mTORC2 signaling and inhibit AKT activation in AML. Blood. 2007;109:3509–3512.
    1. Sarbassov DD, Ali SM, Sengupta S, Sheen JH, Hsu PP, et al. Prolonged rapamycin treatment inhibits mTORC2 assembly and Akt/PKB. Mol Cell. 2006;22:159–168.
    1. Mammucari C, Milan G, Romanello V, Masiero E, Rudolf R, et al. FoxO3 controls autophagy in skeletal muscle in vivo. Cell Metab. 2007;6:458–471.
    1. Rubinsztein DC, Gestwicki JE, Murphy LO, Klionsky DJ. Potential therapeutic applications of autophagy. Nat Rev Drug Discov. 2007;6:304–312.
    1. Yu WH, Cuervo AM, Kumar A, Peterhoff CM, Schmidt SD, et al. Macroautophagy–a novel Beta-amyloid peptide-generating pathway activated in Alzheimer's disease. J Cell Biol. 2005;171:87–98.
    1. Jaeger PA, Wyss-Coray T. All-you-can-eat: autophagy in neurodegeneration and neuroprotection. Mol Neurodegener. 2009;4:16.
    1. Ravikumar B, Vacher C, Berger Z, Davies JE, Luo S, et al. Inhibition of mTOR induces autophagy and reduces toxicity of polyglutamine expansions in fly and mouse models of Huntington disease. Nat Genet. 2004;36:585–595.
    1. Bjorkoy G, Lamark T, Brech A, Outzen H, Perander M, et al. p62/SQSTM1 forms protein aggregates degraded by autophagy and has a protective effect on huntingtin-induced cell death. J Cell Biol. 2005;171:603–614.
    1. Aguib Y, Heiseke A, Gilch S, Riemer C, Baier M, et al. Autophagy induction by trehalose counteracts cellular prion infection. Autophagy. 2009;5:361–369.
    1. Heiseke A, Aguib Y, Riemer C, Baier M, Schatzl HM. Lithium induces clearance of protease resistant prion protein in prion-infected cells by induction of autophagy. J Neurochem. 2009;109:25–34.
    1. Webb JL, Ravikumar B, Atkins J, Skepper JN, Rubinsztein DC. Alpha-Synuclein is degraded by both autophagy and the proteasome. J Biol Chem. 2003;278:25009–25013.
    1. Pickford F, Masliah E, Britschgi M, Lucin K, Narasimhan R, et al. The autophagy-related protein beclin 1 shows reduced expression in early Alzheimer disease and regulates amyloid beta accumulation in mice. J Clin Invest. 2008;118:2190–2199.
    1. Nixon RA. Autophagy, amyloidogenesis and Alzheimer disease. J Cell Sci. 2007;120:4081–4091.
    1. Koo EH, Squazzo SL. Evidence that production and release of amyloid beta-protein involves the endocytic pathway. J Biol Chem. 1994;269:17386–17389.
    1. Roberson ED, Scearce-Levie K, Palop JJ, Yan F, Cheng IH, et al. Reducing endogenous tau ameliorates amyloid beta-induced deficits in an Alzheimer's disease mouse model. Science. 2007;316:750–754.
    1. Myskiw JC, Rossato JI, Bevilaqua LR, Medina JH, Izquierdo I, et al. On the participation of mTOR in recognition memory. Neurobiol Learn Mem. 2008;89:338–351.
    1. Galvan V, Zhang J, Gorostiza OF, Banwait S, Huang W, et al. Long-term prevention of Alzheimer's disease-like behavioral deficits in PDAPP mice carrying a mutation in Asp664. Behav Brain Res. 2008;191:246–255.
    1. Saganich MJ, Schroeder BE, Galvan V, Bredesen DE, Koo EH, et al. Deficits in synaptic transmission and learning in amyloid precursor protein (APP) transgenic mice require C-terminal cleavage of APP. J Neurosci. 2006;26:13428–13436.
    1. Morris R. Developments of a water-maze procedure for studying spatial learning in the rat. J Neurosci Methods. 1984;11:47–60.
    1. Zhang J, Gorostiza OF, Tang H, Bredesen DE, Galvan V. Reversal of learning deficits in hAPP transgenic mice carrying a mutation at Asp664: a role for early experience. Behav Brain Res. 2010;206:202–207.
    1. Meilandt WJ, Cisse M, Ho K, Wu T, Esposito LA, et al. Neprilysin overexpression inhibits plaque formation but fails to reduce pathogenic Abeta oligomers and associated cognitive deficits in human amyloid precursor protein transgenic mice. J Neurosci. 2009;29:1977–1986.
    1. Brown EJ, Albers MW, Shin TB, Ichikawa K, Keith CT, et al. A mammalian protein targeted by G1-arresting rapamycin-receptor complex. Nature. 1994;369:756–758.
    1. Sabatini DM, Erdjument-Bromage H, Lui M, Tempst P, Snyder SH. RAFT1: a mammalian protein that binds to FKBP12 in a rapamycin-dependent fashion and is homologous to yeast TORs. Cell. 1994;78:35–43.
    1. Qiu WQ, Walsh DM, Ye Z, Vekrellis K, Zhang J, et al. Insulin-degrading enzyme regulates extracellular levels of amyloid beta-protein by degradation. J Biol Chem. 1998;273:32730–32738.
    1. Menzies FM, Ravikumar B, Rubinsztein DC. Protective roles for induction of autophagy in multiple proteinopathies. Autophagy. 2006;2:224–225.
    1. Diaz-Troya S, Perez-Perez ME, Florencio FJ, Crespo JL. The role of TOR in autophagy regulation from yeast to plants and mammals. Autophagy. 2008;4:851–865.
    1. Spencer B, Potkar R, Trejo M, Rockenstein E, Patrick C, et al. Beclin 1 gene transfer activates autophagy and ameliorates the neurodegenerative pathology in alpha-synuclein models of Parkinson's and Lewy body diseases. J Neurosci. 2009;29:13578–13588.
    1. Kabeya Y, Mizushima N, Ueno T, Yamamoto A, Kirisako T, et al. LC3, a mammalian homologue of yeast Apg8p, is localized in autophagosome membranes after processing. EMBO J. 2000;19:5720–5728.
    1. Rubinsztein DC, Cuervo AM, Ravikumar B, Sarkar S, Korolchuk V, et al. In search of an “autophagomometer”. Autophagy. 2009;5:585–589.
    1. Tanida I, Minematsu-Ikeguchi N, Ueno T, Kominami E. Lysosomal turnover, but not a cellular level, of endogenous LC3 is a marker for autophagy. Autophagy. 2005;1:84–91.
    1. Bjorkoy G, Lamark T, Pankiv S, Overvatn A, Brech A, et al. Monitoring autophagic degradation of p62/SQSTM1. Methods Enzymol. 2009;452:181–197.
    1. Hansen M, Chandra A, Mitic LL, Onken B, Driscoll M, et al. A role for autophagy in the extension of lifespan by dietary restriction in C. elegans. PLoS Genet. 2008;4:e24.
    1. Vellai T, Takacs-Vellai K, Sass M, Klionsky DJ. The regulation of aging: does autophagy underlie longevity? Trends Cell Biol. 2009;19:487–494.
    1. Cuervo AM, Dice JF. Age-related decline in chaperone-mediated autophagy. J Biol Chem. 2000;275:31505–31513.
    1. Roskam S, Neff F, Schwarting R, Bacher M, Dodel R. APP transgenic mice: the effect of active and passive immunotherapy in cognitive tasks. Neurosci Biobehav Rev. 2010;34:487–499.
    1. Chin J, Massaro CM, Palop JJ, Thwin MT, Yu GQ, et al. Reelin depletion in the entorhinal cortex of human amyloid precursor protein transgenic mice and humans with Alzheimer's disease. J Neurosci. 2007;27:2727–2733.

Source: PubMed

3
Subscribe