Metformin promotes tau aggregation and exacerbates abnormal behavior in a mouse model of tauopathy

Erica Barini, Odetta Antico, Yingjun Zhao, Francesco Asta, Valter Tucci, Tiziano Catelani, Roberto Marotta, Huaxi Xu, Laura Gasparini, Erica Barini, Odetta Antico, Yingjun Zhao, Francesco Asta, Valter Tucci, Tiziano Catelani, Roberto Marotta, Huaxi Xu, Laura Gasparini

Abstract

Background: Alzheimer disease (AD) and other tauopathies develop cerebral intracellular inclusions of hyperphosphorylated tau. Epidemiological and experimental evidence suggests a clear link between type 2 diabetes mellitus and AD. In AD animal models, tau pathology is exacerbated by metabolic comorbidities, such as insulin resistance and diabetes. Within this context, anitidiabetic drugs, including the widely-prescribed insulin-sensitizing drug metformin, are currently being investigated for AD therapy. However, their efficacy for tauopathy in vivo has not been tested.

Results: Here, we report that in the P301S mutant human tau (P301S) transgenic mouse model of tauopathy, chronic administration of metformin exerts paradoxical effects on tau pathology. Despite reducing tau phosphorylation in the cortex and hippocampus via AMPK/mTOR and PP2A, metformin increases insoluble tau species (including tau oligomers) and the number of inclusions with β-sheet aggregates in the brain of P301S mice. In addition, metformin exacerbates hindlimb atrophy, increases P301S hyperactive behavior, induces tau cleavage by caspase 3 and disrupts synaptic structures.

Conclusions: These findings indicate that metformin pro-aggregation effects mitigate the potential benefits arising from its dephosphorylating action, possibly leading to an overall increase of the risk of tauopathy in elderly diabetic patients.

Figures

Fig. 1
Fig. 1
P301S mice show neither peripheral nor brain insulin resistance. a Intraperitoneal glucose tolerance test (IPGTT) in 5 month old WT non-transgenic and P301S transgenic mice. At time 0, P = 0.9; 15 min, P = 0.29; 30 min, P = 0.33; 60 min, P = 0.3; 120 min, P = 0.26; two-way ANOVA followed by Holm-Sidak multiple comparison test. P301S, n = 10; WT, n = 12. b IPGTT AUC. P = 0.17; Student t-test. c Insulin tolerance test (ITT) in 5 month old WT non-transgenic and P301S transgenic mice. At time 0, P = 0.09; 15 min, P = 0.95, 30 min, P = 0.69; 45 min, P = 0.73; 60 min, P = 0.24; 90 min, P = 0.11; 120 min, P = 0.22; two-way ANOVA followed by Holm-Sidak multiple comparison test. P301S, n = 12; WT, n = 6. In a and c, data points represent the average glycemia ± SEM. d ITT AUC. P = 0.50, Student t-test. e-f Glucose concentration (e) and insulin levels (f) in the cortex of 5-month old P301S and WT mice. n = 6 / group. Glucose and insulin concentrations were normalized to the sample protein content. Bars represent the average ± SEM. In panel e, P = 0.79; panel f, P = 0.24; Student t-test. g Western blot of mTOR, IRβ and Akt and total tau in the cortex. Actin and Tubulin were analyzed as a loading control. h Quantitative analysis of protein expression of IRβ (WT, n = 9; P301S, n = 12), AKT (WT, n = 8; P301S, n = 12) and mTOR (WT, n = 6; P301S, n = 7) in the cortex of 5-month old P301S and WT mice. Protein levels were normalized to actin. Data are expressed as percentage of WT. Bars represent the average ± SEM. IRβ, P = 0.06; AKT, P = 0.09; mTOR, P = 0.14; Student t-test
Fig. 2
Fig. 2
Metformin treatment reduces tau phosphorylation in vivo. Total tau and phosphorylated tau (pTau) were analyzed in the cortex and hippocampus of 5 month old P301S mice treated with or without metformin for 4 months. a Western blot of phosphorylated and total tau in the cortex. Actin was analyzed as a loading control. b-g Quantitative analysis of pTau in the cortex (b, d, f, g) and hippocampus (c, e). Phosphorylated tau was analyzed using AT8 (b, c), anti-phospho-S262 tau (d, e), PHF-1 (f), AT270 (g) antibodies and normalized to total tau levels. Total tau was analyzed using Tau5 antibody. Bars represent the average ratio over total tau ± SEM. p < 0.05, Student t-test. Panel b: n = 11 / group. Panel c: Ctr, n = 10; Met, n = 11. Panel d: Ctr, n = 12; Met, n = 12. Panel e: Ctr, n = 7; Met, n = 6; Panel f-g: Ctr, n = 7; Met, n = 7. h-i Representative fluorescence images of AT8 immunoreactivity in the cortex of untreated and metformin-treated P301S mice. Scale bars: in H-I, 50 μm; in h’-i’, 150 μm. Panels h’ and i’ are magnification of squared areas in panels h and i. j. AT8 positive (AT8+) cells in the prefrontal cortex. k-l Representative fluorescence images of AT8 immunoreactivity in hippocampi of untreated and metformin-treated P301S mice. Scale bars: in k-l, 100 μm; in k’-l’, 200 μm. Panels k’ and l’ are magnification of squared areas in panels k and l, respectively. m-o AT8 positive (AT8+) cells in the hippocampus (m), granular zone (n) and dentate gyrus (o). Panel j: Ctr, n = 3; Met, n = 4; Panel m-o: Ctr, n = 3; Met, n = 5. In j and m-o, bars represent the average cell number / mm2 ± SEM. *p < 0.05, **p < 0.01, Student t-test
Fig. 3
Fig. 3
Metformin treatment induces PP2A expression, inhibits mTOR and activates AMPK in the brain of P301S mice. Protein levels of PP2A, GSK3β, pGSK3β, mTOR, S6, phospho-S6 (pS6), AMPK and phospho-AMPK (pAMPK) were analyzed by western blot in the brain of 5 month old P301S mice treated with or without metformin for 4 months. a Western blot of PP2A in the cortex. Tubulin was analyzed as a loading control. b-c Quantitative analysis of PP2A in the cortex (b) and hippocampus (c). In panel b: Ctr, n = 10; Met, n = 12. In panel c: Ctr, n = 9; Met, n = 11. d-e Quantitative analysis of p[S9]GSK3β (d) and GSK3β (e) in the cortex. Ctr, n = 13; Met, n = 12. f Western blot of mTOR in the cortex. Actin was analyzed as a loading control. g Quantitative analysis of mTOR protein expression. Ctr, n = 7; Met, n = 7. h Western blot of pS6 and S6 in the cortex. Tubulin was analyzed as a loading control. i-j Quantitative analysis of pS6 (i) and S6 (j). n = 7 / group. k Western blot of pAMPK and AMPK in the cortex. Tubulin was analyzed as a loading control. l-m Quantitative analysis of AMPK (l) and pAMPK (m). Panel l-m: Ctr, n = 10; Met, n = 12. Protein expression levels were normalized to tubulin or actin as indicated in the graphs. Levels of phosphorylated proteins were normalized to the total respective protein. In all graphs, bars represent the average ratio ± SEM. *p < 0.05, **p < 0.01, Student t-test
Fig. 4
Fig. 4
Metformin reduces tau phosphorylation in mouse cortical neurons via the AMPK-mTOR pathway. Primary cortical neurons were incubated for 6 h with or without 2.5 mM metformin (Met), 10 nM okadaic acid (OA), 10 μM rapamycin (Rapa), and 10 μM dorsomorphin (Dorso) alone or in combination. All treatments were performed in the absence of insulin, except when specifically indicated. a Quantitative analysis of phospho-tau (pTau) in cortical neurons treated with or without metformin in the absence or presence of insulin. b Quantitative analysis of pTau in cortical neurons treated with or without metformin in the absence or presence of okadaic acid. c-d Quantitative analysis of pAMPK (c) and pTau (d) in cortical neurons treated with or without metformin and dorsomorphin alone or in combination. e Quantitative analysis of pS6 in cortical neurons treated with or without metformin in the absence or presence of insulin. f-g. Quantitative analysis of pTau (f) and pS6 (g) in cortical neurons treated with or without metformin and rapamycin. h-i. Quantitative analysis of pS6 (h) and pTau (i) in neurons non-infected (NI), infected with GFP and S16H-Rheb or GFP alone and treated with metformin. Levels of phosphorylated protein were normalized to levels of respective proteins. In panels a, b, d, f, and i, pTau and total tau were analyzed using AT8 and Tau5 antibodies, respectively. In all graphs, bars represent the average ratio ± SEM of 3 independent experiments. *p < 0.05, **p < 0.01, one-way ANOVA followed by Holm-Sidak multiple comparison test
Fig. 5
Fig. 5
Metformin increases the number of tau inclusions in P301S mouse brain in vivo and induces aggregation of recombinant P301S tau in vitro. a Western blot analysis of tau in the sarkosyl-insoluble cortical fraction from P301S mice treated with or without metformin. The arrow indicates the bands of tau dimers. Tau was detected using the phosphorylation-and conformation dependent AT100 and Tau5 anti-tau monoclonal antibodies. MemCode was used to control equal loading. b-c Quantitative analysis of monomeric (b) and dimeric (c) tau in sarkosyl-insoluble fractions of P301S cortex. Densitometric values of tau bands are normalized over MemCode and bars represent the average ratio ± SEM. P301S, n = 4 / group. d. Representative images of MC1 immunoreactivity (d, d’; green) and FSB (d, d”; light blue) fluorescence in the cortex of P301S mice. Scale bars: in d, 50 μm; in d’-d”, 100 μm. e Representative images of AT8 immunoreactivity (e, e’; green) and FSB (e, e”; light blue) fluorescence in the cortex of P301S mice. Scale bars: in e, 50 μm; in e’-e”, 150 μm. f-g FSB+ cells in the prefrontal cortex (f) and hippocampus (g) of P301S mice. n = 3 / group. Bars represent the average number of FSB+ cells / mm2 ± SEM. **p < 0.01, Student t-test. h-i. Gallyas silver staining in the cortex of P301S mice untreated (h) or treated with metformin (i). Scale bar 150 μm. j-k. Gallyas+ cells in the prefrontal cortex (j) and hippocampus (k) of P301S mice. n = 3 / group. Bars represent the average number of Gallyas+ cells / mm2 ± SEM. **p < 0.01, Student t-test. l. Thioflavin T (ThT) fluorescence of recombinant P301S mutant human tau (P301Stau) aggregated in vitro in the absence (Ctr) or presence of 37.5 μM heparin (Hep) and/or 30 μM metformin (Met) for 2, 7 and 14 days. Data points represent the average ± SEM from at least 3 independent experiments. m-n. Representative negative staining electron microscopy image of fibrils of recombinant P301Stau aggregated in vitro for 7 days in presence of heparin (m) or metformin (n). In m, inset shows a high magnification of the squared area. In n, inset shows a high magnification image of tau aggregates. Scale bars: i-j, 2.5 μm; insets, 0.5 μm. o ThT fluorescence of recombinant P301Stau aggregated in vitro in the absence (control, Ctr) or presence of 37.5 μM heparin, 30 μM metformin, 30 μM phenformin (Phe) or 30 μM buformin (Buf) for 7 days. Fluorescence values are expressed as arbitrary units and normalized to the control. Bars represent the average ± SEM. **p < 0.01, one-way ANOVA followed by Holm-Sidak multiple comparison test. p Quantitative analysis of pTau in cortical neurons treated with or without 2.5 mM metformin, 30 μM phenformin and 30 μM buformin. pTau was detected using the AT8 monoclonal antibody and normalized on total tau detected by Tau5 antibody. Data are expressed as percentage of Ctr. Bars represent the average percentage ± SEM from 2 independent experiments. *p < 0.05, **p < 0.01, one-way ANOVA followed by Holm-Sidak multiple comparison test
Figure 6
Figure 6
Metformin worsens hindlimb atrophy and hyperactive behavior and induces caspase 3 activation and tau cleavage. a-c Open field test in 5-month old P301S transgenic mice and age-matched WT non transgenic mice. Distance travelled (a), speed (b) and number of rearings (c) are shown. Bars represent the average values ± SEM. *p < 0.05, Student t-test. n = 9 / group. d-g, k-s. P301S mice were treated with or without metformin (Met) for 4 months starting from 1 month of age. d-f. Open field test. Distance travelled (d), speed (e) and rearing (f) are shown. Bars represent the average values ± SEM. *p < 0.05, Student t-test. Panels d-f: Ctr, n = 11; Met, n = 9. g Hind limb extension test in P301S mice treated with or without metformin. Data points represent the average score ± SEM. *p < 0.05, two-way ANOVA followed by Holm-Sidak multiple comparison test. A.U., arbitrary units. Ctr, n = 12; Met, n = 13. h-j. Primary cortical neurons were treated with or without 0.5 mM metformin for 24 h. h-i. Western blot (h) and quantitative analyses (i) of cleaved caspase 3 (c-caspase 3), c-Tau, c-PARP and PSD95. Protein levels were normalized to actin. Bars represent the average values ± SEM. *p < 0.05, **p < 0.01, Student t-test. n = 3/group. j. Quantification of clusters with overlapping synapsin-I and PSD95 immunoreactivity. Bars represent the average ± SEM. **p < 0.01, Student t-test. n = 11/group. k Representative confocal maximal projection images of active c-caspase 3 immunoreactivity (green) in the prefrontal cortex of untreated and metformin-treated P301S mice. Nuclei are counterstained with Hoechst 33342. Scale bar 10 μm. l Quantitative analysis of active c-caspase 3. c-caspase 3 was analyzed by western blot and protein levels were normalized to tubulin. Bars represent the average ± SEM. n = 7 / group. *p < 0.05, Student t-test. m Western blot analysis of cleaved-tau (c-Tau) in the soluble and sarkosyl-insoluble fraction of metformin-treated and untreated P301S mouse cortex. n-o Quantitative analysis of cleaved Tau (c-Tau) in the soluble (n) and insoluble (o) cortical fraction. c-Tau was analyzed by western blot and protein levels were normalized to actin (n) or MemCode (o). Bars represent the average ± SEM. Panel n: n = 7 / group. Panel o: n = 3/group. p Western blot analysis of synapsin I, PSD95, synaptophysin, tubulin and GAPDH in the cortex of P301S mice treated with or without metformin. q-s. Quantitative analysis of synapsin I (q), PSD95 (r) and synaptophysin (s). Protein levels were analyzed by western bot and normalized to tubulin or GAPDH. Bars represent the average ± SEM. *p < 0.05, Student t-test. Panel q: n = 7 / group. Panel r: Ctr, n = 13; Met, n = 10. Panel s: Ctr, n = 13; Met, n = 11

References

    1. Bateman RJ, Xiong C, Benzinger TL, Fagan AM, Goate A, Fox NC, et al. Clinical and biomarker changes in dominantly inherited Alzheimer’s disease. N Engl J Med. 2012;367:795–804. doi: 10.1056/NEJMoa1202753.
    1. Arriagada PV, Growdon JH, Hedley-Whyte ET, Hyman BT. Neurofibrillary tangles but not senile plaques parallel duration and severity of Alzheimer’s disease. Neurology. 1992;42:631–9. doi: 10.1212/WNL.42.3.631.
    1. Gasparini L, Netzer WJ, Greengard P, Xu H. Does insulin dysfunction play a role in Alzheimer’s disease? Trends Pharmacol Sci. 2002;23:288–93. doi: 10.1016/S0165-6147(02)02037-0.
    1. El Khoury NB, Gratuze M, Papon MA, Bretteville A, Planel E. Insulin dysfunction and Tau pathology. Front Cell Neurosci. 2014;8:22. doi: 10.3389/fncel.2014.00022.
    1. Cohen AD, Klunk WE. Early detection of Alzheimer’s disease using PiB and FDG PET. Neurobiol Dis. 2014;72 Pt A:117–22. doi: 10.1016/j.nbd.2014.05.001.
    1. Talbot K, Wang HY, Kazi H, Han LY, Bakshi KP, Stucky A, 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. 2012;122:1316–38. doi: 10.1172/JCI59903.
    1. Yarchoan M, Toledo JB, Lee EB, Arvanitakis Z, Kazi H, Han LY, et al. Abnormal serine phosphorylation of insulin receptor substrate 1 is associated with tau pathology in Alzheimer’s disease and tauopathies. Acta Neuropathol. 2014;128:679–89. doi: 10.1007/s00401-014-1328-5.
    1. Lesort M, Jope RS, Johnson GV. Insulin transiently increases tau phosphorylation: involvement of glycogen synthase kinase-3beta and Fyn tyrosine kinase. J Neurochem. 1999;72:576–84. doi: 10.1046/j.1471-4159.1999.0720576.x.
    1. Hong M, Lee VM. Insulin and insulin-like growth factor-1 regulate tau phosphorylation in cultured human neurons. J Biol Chem. 1997;272:19547–53. doi: 10.1074/jbc.272.31.19547.
    1. Gupta A, Bisht B, Dey CS. Peripheral insulin-sensitizer drug metformin ameliorates neuronal insulin resistance and Alzheimer’s-like changes. Neuropharmacology. 2011;60:910–20. doi: 10.1016/j.neuropharm.2011.01.033.
    1. Yarchoan M, Arnold SE. Repurposing diabetes drugs for brain insulin resistance in Alzheimer disease. Diabetes. 2014;63:2253–61. doi: 10.2337/db14-0287.
    1. Pintana H, Apaijai N, Pratchayasakul W, Chattipakorn N, Chattipakorn SC. Effects of metformin on learning and memory behaviors and brain mitochondrial functions in high fat diet induced insulin resistant rats. Life Sci. 2012;91:409–14. doi: 10.1016/j.lfs.2012.08.017.
    1. McNeilly AD, Williamson R, Balfour DJ, Stewart CA, Sutherland C. A high-fat-diet-induced cognitive deficit in rats that is not prevented by improving insulin sensitivity with metformin. Diabetologia. 2012;55:3061–70. doi: 10.1007/s00125-012-2686-y.
    1. Li J, Deng J, Sheng W, Zuo Z. Metformin attenuates Alzheimer’s disease-like neuropathology in obese, leptin-resistant mice. Pharmacol Biochem Behav. 2012;101:564–74. doi: 10.1016/j.pbb.2012.03.002.
    1. Zhao RR, Xu XC, Xu F, Zhang WL, Zhang WL, Liu LM, et al. Metformin protects against seizures, learning and memory impairments and oxidative damage induced by pentylenetetrazole-induced kindling in mice. Biochem Biophys Res Commun. 2014;448:414–7. doi: 10.1016/j.bbrc.2014.04.130.
    1. Adeyemi OO, Ishola IO, Adedeji HA. Novel action of metformin in the prevention of haloperidol-induced catalepsy in mice: Potential in the treatment of Parkinson’s disease? Prog Neuropsychopharmacol Biol Psychiatry 2013. doi: 10.1016/j.pnpbp.2013.10.014. [Epub ahead of print].
    1. Kickstein E, Krauss S, Thornhill P, Rutschow D, Zeller R, Sharkey J, et al. Biguanide metformin acts on tau phosphorylation via mTOR/protein phosphatase 2A (PP2A) signaling. Proc Natl Acad Sci U S A. 2010;107:21830–5. doi: 10.1073/pnas.0912793107.
    1. Allen B, Ingram E, Takao M, Smith MJ, Jakes R, Virdee K, et al. Abundant tau filaments and nonapoptotic neurodegeneration in transgenic mice expressing human P301S tau protein. J Neurosci. 2002;22:9340–51.
    1. Hampton DW, Webber DJ, Bilican B, Goedert M, Spillantini MG, Chandran S. Cell-mediated neuroprotection in a mouse model of human tauopathy. J Neurosci. 2010;30:9973–83. doi: 10.1523/JNEUROSCI.0834-10.2010.
    1. Scattoni ML, Gasparini L, Alleva E, Goedert M, Calamandrei G, Spillantini MG. Early behavioural markers of disease in P301S tau transgenic mice. Behav Brain Res. 2010;208:250–7. doi: 10.1016/j.bbr.2009.12.002.
    1. Delobel P, Lavenir I, Fraser G, Ingram E, Holzer M, Ghetti B, et al. Analysis of tau phosphorylation and truncation in a mouse model of human tauopathy. Am J Pathol. 2008;172:123–31. doi: 10.2353/ajpath.2008.070627.
    1. Tucker GT, Casey C, Phillips PJ, Connor H, Ward JD, Woods HF. Metformin kinetics in healthy subjects and in patients with diabetes mellitus. Br J Clin Pharmacol. 1981;12:235–46. doi: 10.1111/j.1365-2125.1981.tb01206.x.
    1. Liu F, Grundke-Iqbal I, Iqbal K, Gong CX. Contributions of protein phosphatases PP1, PP2A, PP2B and PP5 to the regulation of tau phosphorylation. Eur J Neurosci. 2005;22:1942–50. doi: 10.1111/j.1460-9568.2005.04391.x.
    1. Avila J, Leon-Espinosa G, Garcia E, Garcia-Escudero V, Hernandez F, Defelipe J. Tau phosphorylation by GSK3 in different conditions. Int J Alzheimers Dis. 2012;2012:578373.
    1. Doble BW, Woodgett JR. GSK-3: tricks of the trade for a multi-tasking kinase. J Cell Sci. 2003;116:1175–86. doi: 10.1242/jcs.00384.
    1. Rodgers JT, Vogel RO, Puigserver P. Clk2 and B56beta mediate insulin-regulated assembly of the PP2A phosphatase holoenzyme complex on Akt. Mol Cell. 2011;41:471–9. doi: 10.1016/j.molcel.2011.02.007.
    1. Hartley D, Cooper GM. Role of mTOR in the degradation of IRS-1: regulation of PP2A activity. J Cell Biochem. 2002;85:304–14. doi: 10.1002/jcb.10135.
    1. Zhang HH, Lipovsky AI, Dibble CC, Sahin M, Manning BD. S6K1 regulates GSK3 under conditions of mTOR-dependent feedback inhibition of Akt. Mol Cell. 2006;24:185–97. doi: 10.1016/j.molcel.2006.09.019.
    1. Magnuson B, Ekim B, Fingar DC. Regulation and function of ribosomal protein S6 kinase (S6K) within mTOR signalling networks. Biochem J. 2012;441:1–21. doi: 10.1042/BJ20110892.
    1. Edinger AL, Thompson CB. Akt maintains cell size and survival by increasing mTOR-dependent nutrient uptake. Mol Biol Cell. 2002;13:2276–88. doi: 10.1091/mbc.01-12-0584.
    1. Cheng SW, Fryer LG, Carling D, Shepherd PR. Thr2446 is a novel mammalian target of rapamycin (mTOR) phosphorylation site regulated by nutrient status. J Biol Chem. 2004;279:15719–22. doi: 10.1074/jbc.C300534200.
    1. Hawley SA, Gadalla AE, Olsen GS, Hardie DG. The antidiabetic drug metformin activates the AMP-activated protein kinase cascade via an adenine nucleotide-independent mechanism. Diabetes. 2002;51:2420–5. doi: 10.2337/diabetes.51.8.2420.
    1. Zhou G, Myers R, Li Y, Chen Y, Shen X, Fenyk-Melody J, et al. Role of AMP-activated protein kinase in mechanism of metformin action. J Clin Invest. 2001;108:1167–74. doi: 10.1172/JCI13505.
    1. Yan L, Findlay GM, Jones R, Procter J, Cao Y, Lamb RF. Hyperactivation of mammalian target of rapamycin (mTOR) signaling by a gain-of-function mutant of the Rheb GTPase. J Biol Chem. 2006;281:19793–7. doi: 10.1074/jbc.C600028200.
    1. Augustinack JC, Schneider A, Mandelkow EM, Hyman BT. Specific tau phosphorylation sites correlate with severity of neuronal cytopathology in Alzheimer’s disease. Acta Neuropathol. 2002;103:26–35. doi: 10.1007/s004010100423.
    1. Dickson DW, Crystal HA, Bevona C, Honer W, Vincent I, Davies P. Correlations of synaptic and pathological markers with cognition of the elderly. Neurobiol Aging. 1995;16:285–98. doi: 10.1016/0197-4580(95)00013-5.
    1. Xu S, Brunden KR, Trojanowski JQ, Lee VM. Characterization of tau fibrillization in vitro. Alzheimers Dement. 2010;6:110–7. doi: 10.1016/j.jalz.2009.06.002.
    1. Pezze M, McGarrity S, Mason R, Fone KC, Bast T. Too little and too much: hypoactivation and disinhibition of medial prefrontal cortex cause attentional deficits. J Neurosci. 2014;34:7931–46. doi: 10.1523/JNEUROSCI.3450-13.2014.
    1. Bardgett ME, Baum KT, O’Connell SM, Lee NM, Hon JC. Effects of risperidone on locomotor activity and spatial memory in rats with hippocampal damage. Neuropharmacology. 2006;51:1156–62. doi: 10.1016/j.neuropharm.2006.07.014.
    1. Barneoud P, Lolivier J, Sanger DJ, Scatton B, Moser P. Quantitative motor assessment in FALS mice: a longitudinal study. Neuroreport. 1997;8:2861–5. doi: 10.1097/00001756-199709080-00012.
    1. Gamblin TC, Chen F, Zambrano A, Abraha A, Lagalwar S, Guillozet AL, et al. Caspase cleavage of tau: linking amyloid and neurofibrillary tangles in Alzheimer’s disease. Proc Natl Acad Sci U S A. 2003;100:10032–7. doi: 10.1073/pnas.1630428100.
    1. Rissman RA, Poon WW, Blurton-Jones M, Oddo S, Torp R, Vitek MP, et al. Caspase-cleavage of tau is an early event in Alzheimer disease tangle pathology. J Clin Invest. 2004;114:121–30. doi: 10.1172/JCI200420640.
    1. de Calignon A, Fox LM, Pitstick R, Carlson GA, Bacskai BJ, Spires-Jones TL, et al. Caspase activation precedes and leads to tangles. Nature. 2010;464:1201–4. doi: 10.1038/nature08890.
    1. Zhao Y, Tseng IC, Heyser CJ, Rockenstein E, Mante M, Adame A, et al. Appoptosin-mediated caspase cleavage of tau contributes to progressive supranuclear palsy pathogenesis. Neuron. 2015;87:963–75. doi: 10.1016/j.neuron.2015.08.020.
    1. Bomfim TR, Forny-Germano L, Sathler LB, Brito-Moreira J, Houzel JC, Decker H, et al. An anti-diabetes agent protects the mouse brain from defective insulin signaling caused by Alzheimer’s disease- associated Abeta oligomers. J Clin Invest. 2012;122:1339–53. doi: 10.1172/JCI57256.
    1. Zhao WQ, De Felice FG, Fernandez S, Chen H, Lambert MP, Quon MJ, et al. Amyloid beta oligomers induce impairment of neuronal insulin receptors. FASEB J. 2008;22:246–60. doi: 10.1096/fj.06-7703com.
    1. Viollet B, Guigas B, Sanz Garcia N, Leclerc J, Foretz M, Andreelli F. Cellular and molecular mechanisms of metformin: an overview. Clin Sci. 2012;122:253–70. doi: 10.1042/CS20110386.
    1. Hundal RS, Krssak M, Dufour S, Laurent D, Lebon V, Chandramouli V, et al. Mechanism by which metformin reduces glucose production in type 2 diabetes. Diabetes. 2000;49:2063–9. doi: 10.2337/diabetes.49.12.2063.
    1. Cabreiro F, Au C, Leung KY, Vergara-Irigaray N, Cocheme HM, Noori T, et al. Metformin retards aging in C. elegans by altering microbial folate and methionine metabolism. Cell. 2013;153:228–39. doi: 10.1016/j.cell.2013.02.035.
    1. De Haes W, Frooninckx L, Van Assche R, Smolders A, Depuydt G, Billen J, et al. Metformin promotes lifespan through mitohormesis via the peroxiredoxin PRDX-2. Proc Natl Acad Sci U S A. 2014;111:E2501–9. doi: 10.1073/pnas.1321776111.
    1. Anisimov VN, Berstein LM, Egormin PA, Piskunova TS, Popovich IG, Zabezhinski MA, et al. Metformin slows down aging and extends life span of female SHR mice. Cell Cycle. 2008;7:2769–73. doi: 10.4161/cc.7.17.6625.
    1. Martin-Montalvo A, Mercken EM, Mitchell SJ, Palacios HH, Mote PL, Scheibye-Knudsen M, et al. Metformin improves healthspan and lifespan in mice. Nat Commun. 2013;4:2192. doi: 10.1038/ncomms3192.
    1. Hayden EC. Anti-Ageing pill pushed as bona fide drug. Nature. 2015;522:265–6. doi: 10.1038/522265a.
    1. Ozcelik S, Fraser G, Castets P, Schaeffer V, Skachokova Z, Breu K, et al. Rapamycin attenuates the progression of tau pathology in P301S tau transgenic mice. PLoS ONE. 2013;8:e62459. doi: 10.1371/journal.pone.0062459.
    1. Caccamo A, Magri A, Medina DX, Wisely EV, Lopez-Aranda MF, Silva AJ, et al. mTOR regulates tau phosphorylation and degradation: implications for Alzheimer’s disease and other tauopathies. Aging Cell. 2013;12:370–80. doi: 10.1111/acel.12057.
    1. Orr ME, Salinas A, Buffenstein R, Oddo S. Mammalian target of rapamycin hyperactivity mediates the detrimental effects of a high sucrose diet on Alzheimer’s disease pathology. Neurobiol Aging. 2014;35:1233–42. doi: 10.1016/j.neurobiolaging.2013.12.006.
    1. Caccamo A, Majumder S, Richardson A, Strong R, Oddo S. Molecular interplay between mammalian target of rapamycin (mTOR), amyloid-beta, and Tau: effects on cognitive impairments. J Biol Chem. 2010;285:13107–20. doi: 10.1074/jbc.M110.100420.
    1. van Weeren PC, de Bruyn KM, de Vries-Smits AM, van Lint J, Burgering BM. Essential role for protein kinase B (PKB) in insulin-induced glycogen synthase kinase 3 inactivation. Characterization of dominant-negative mutant of PKB. J Biol Chem. 1998;273:13150–6. doi: 10.1074/jbc.273.21.13150.
    1. Maeda S, Sahara N, Saito Y, Murayama M, Yoshiike Y, Kim H, et al. Granular tau oligomers as intermediates of tau filaments. Biochemistry. 2007;46:3856–61. doi: 10.1021/bi061359o.
    1. Ma TC, Buescher JL, Oatis B, Funk JA, Nash AJ, Carrier RL, et al. Metformin therapy in a transgenic mouse model of Huntington’s disease. Neurosci Lett. 2007;411:98–103. doi: 10.1016/j.neulet.2006.10.039.
    1. Moore EM, Mander AG, Ames D, Kotowicz MA, Carne RP, Brodaty H, et al. Increased risk of cognitive impairment in patients with diabetes is associated with metformin. Diabetes Care. 2013;36:2981–7. doi: 10.2337/dc13-0229.
    1. Imfeld P, Bodmer M, Jick SS, Meier CR. Metformin, other antidiabetic drugs, and risk of Alzheimer’s disease: a population-based case-control study. J Am Geriatr Soc. 2012;60:916–21. doi: 10.1111/j.1532-5415.2012.03916.x.
    1. Chen Y, Zhou K, Wang R, Liu Y, Kwak YD, Ma T, et al. Antidiabetic drug metformin (GlucophageR) increases biogenesis of Alzheimer’s amyloid peptides via up-regulating BACE1 transcription. Proc Natl Acad Sci U S A. 2009;106:3907–12. doi: 10.1073/pnas.0807991106.
    1. Chai X, Wu S, Murray TK, Kinley R, Cella CV, Sims H, et al. Passive immunization with anti-Tau antibodies in two transgenic models: reduction of Tau pathology and delay of disease progression. J Biol Chem. 2011;286:34457–67. doi: 10.1074/jbc.M111.229633.
    1. Geda YE, Schneider LS, Gitlin LN, Miller DS, Smith GS, Bell J, et al. Neuropsychiatric symptoms in Alzheimer’s disease: past progress and anticipation of the future. Alzheimers Dement. 2013;9:602–8. doi: 10.1016/j.jalz.2012.12.001.
    1. SantaCruz K, Lewis J, Spires T, Paulson J, Kotilinek L, Ingelsson M, et al. Tau suppression in a neurodegenerative mouse model improves memory function. Science. 2005;309:476–81. doi: 10.1126/science.1113694.
    1. Hampp C, Borders-Hemphill V, Moeny DG, Wysowski DK. Use of antidiabetic drugs in the U.S., 2003–2012. Diabetes Care. 2014;37:1367–74. doi: 10.2337/dc13-2289.
    1. Hebert LE, Weuve J, Scherr PA, Evans DA. Alzheimer disease in the United States (2010–2050) estimated using the 2010 census. Neurology. 2013;80:1778–83. doi: 10.1212/WNL.0b013e31828726f5.
    1. Wild S, Roglic G, Green A, Sicree R, King H. Global prevalence of diabetes: estimates for the year 2000 and projections for 2030. Diabetes Care. 2004;27:1047–53. doi: 10.2337/diacare.27.5.1047.
    1. Otvos L, Jr, Feiner L, Lang E, Szendrei GI, Goedert M, Lee VM. Monoclonal antibody PHF-1 recognizes tau protein phosphorylated at serine residues 396 and 404. J Neurosci Res. 1994;39:669–73. doi: 10.1002/jnr.490390607.
    1. Labuzek K, Suchy D, Gabryel B, Bielecka A, Liber S, Okopien B. Quantification of metformin by the HPLC method in brain regions, cerebrospinal fluid and plasma of rats treated with lipopolysaccharide. Pharmacol Rep. 2010;62:956–65. doi: 10.1016/S1734-1140(10)70357-1.
    1. Ferrera D, Mazzaro N, Canale C, Gasparini L. Resting microglia react to A beta 42 fibrils but do not detect oligomers or oligomer-induced neuronal damage. Neurobiol Aging. 2014;35:2444–57. doi: 10.1016/j.neurobiolaging.2014.05.023.
    1. Nie D, Di Nardo A, Han JM, Baharanyi H, Kramvis I, Huynh T, et al. Tsc2-Rheb signaling regulates EphA-mediated axon guidance. Nat Neurosci. 2010;13:163–72. doi: 10.1038/nn.2477.
    1. Mostoslavsky G, Kotton DN, Fabian AJ, Gray JT, Lee JS, Mulligan RC. Efficiency of transduction of highly purified murine hematopoietic stem cells by lentiviral and oncoretroviral vectors under conditions of minimal in vitro manipulation. Mol Ther. 2005;11:932–40. doi: 10.1016/j.ymthe.2005.01.005.
    1. Naldini L, Blomer U, Gallay P, Ory D, Mulligan R, Gage FH, et al. In vivo gene delivery and stable transduction of nondividing cells by a lentiviral vector. Science. 1996;272:263–7. doi: 10.1126/science.272.5259.263.
    1. Gasparini L, Crowther RA, Martin KR, Berg N, Coleman M, Goedert M, et al. Tau inclusions in retinal ganglion cells of human P301S tau transgenic mice: Effects on axonal viability. Neurobiol Aging. 2011;32:419–33. doi: 10.1016/j.neurobiolaging.2009.03.002.
    1. Schneider CA, Rasband WS, Eliceiri KW. NIH Image to ImageJ: 25 years of image analysis. Nat Methods. 2012;9:671–5. doi: 10.1038/nmeth.2089.
    1. Sato K, Higuchi M, Iwata N, Saido TC, Sasamoto K. Fluoro-substituted and 13C-labeled styrylbenzene derivatives for detecting brain amyloid plaques. Eur J Med Chem. 2004;39:573–8. doi: 10.1016/j.ejmech.2004.02.013.
    1. Hasegawa M, Smith MJ, Goedert M. Tau proteins with FTDP-17 mutations have a reduced ability to promote microtubule assembly. FEBS Lett. 1998;437:207–10. doi: 10.1016/S0014-5793(98)01217-4.

Source: PubMed

Подписаться