Tauopathy induced by low level expression of a human brain-derived tau fragment in mice is rescued by phenylbutyrate

Marie K Bondulich, Tong Guo, Christopher Meehan, John Manion, Teresa Rodriguez Martin, Jacqueline C Mitchell, Tibor Hortobagyi, Natalia Yankova, Virginie Stygelbout, Jean-Pierre Brion, Wendy Noble, Diane P Hanger, Marie K Bondulich, Tong Guo, Christopher Meehan, John Manion, Teresa Rodriguez Martin, Jacqueline C Mitchell, Tibor Hortobagyi, Natalia Yankova, Virginie Stygelbout, Jean-Pierre Brion, Wendy Noble, Diane P Hanger

Abstract

Human neurodegenerative tauopathies exhibit pathological tau aggregates in the brain along with diverse clinical features including cognitive and motor dysfunction. Post-translational modifications including phosphorylation, ubiquitination and truncation, are characteristic features of tau present in the brain in human tauopathy. We have previously reported an N-terminally truncated form of tau in human brain that is associated with the development of tauopathy and is highly phosphorylated. We have generated a new mouse model of tauopathy in which this human brain-derived, 35 kDa tau fragment (Tau35) is expressed in the absence of any mutation and under the control of the human tau promoter. Most existing mouse models of tauopathy overexpress mutant tau at levels that do not occur in human neurodegenerative disease, whereas Tau35 transgene expression is equivalent to less than 10% of that of endogenous mouse tau. Tau35 mice recapitulate key features of human tauopathies, including aggregated and abnormally phosphorylated tau, progressive cognitive and motor deficits, autophagic/lysosomal dysfunction, loss of synaptic protein, and reduced life-span. Importantly, we found that sodium 4-phenylbutyrate (Buphenyl®), a drug used to treat urea cycle disorders and currently in clinical trials for a range of neurodegenerative diseases, reverses the observed abnormalities in tau and autophagy, behavioural deficits, and loss of synapsin 1 in Tau35 mice. Our results show for the first time that, unlike other tau transgenic mouse models, minimal expression of a human disease-associated tau fragment in Tau35 mice causes a profound and progressive tauopathy and cognitive changes, which are rescued by pharmacological intervention using a clinically approved drug. These novel Tau35 mice therefore represent a highly disease-relevant animal model in which to investigate molecular mechanisms and to develop novel treatments for human tauopathies.

Keywords: 4-phenylbutyrate; lysosomal degradation; progressive supranuclear palsy; tau; tauopathies.

© The Author (2016). Published by Oxford University Press on behalf of the Guarantors of Brain.

Figures

https://www.ncbi.nlm.nih.gov/pmc/articles/instance/4958900/bin/aww137fig1g.jpg
Post-translational modification of tau is common in human tauopathies. Bondulich et al. generate transgenic mice expressing low levels of a truncated form of tau (Tau35) that is associated with human tauopathy. Tau35 mice develop progressive tau neuropathology and cognitive impairment, modelling human disease. The approved drug 4-phenylbutyrate rescues these abnormalities.
Figure 1
Figure 1
Tau expression in human tauopathy and Tau35 mouse brain. (A) Construct used to generate Tau35 mice with the human tau promoter (phTau), upstream of the Tau35 sequence with the haemagglutinin tag (HA). The hypoxanthine phosphoribosyltransferase promoter (pHprt) and exons 1–3 enabled targeted integration of the Tau35-HA transgene. The lower panel shows a schematic representation of the expressed Tau35-HA protein in comparison to full-length human tau (441 amino acids). The amino terminal domain of tau contains two inserts (N1, N2), followed by a central proline-rich domain and the microtubule (MT) binding domain, which comprises four repeats (R1-R4). Tau35 retains the majority of the proline-rich domain, four MT binding repeats and an intact C-terminus. The epitopes of the phospho-dependent (orange boxes), conformation-dependent (black boxes) and region-specific (grey boxes) tau antibodies used in this study are indicated above full-length tau. (B) Western blot of insoluble fractions of control and progressive supranuclear palsy (PSP) brain reveals the 35 kDa tau species (asterisk) in the premotor cortex of human PSP brain probed with TP70, the C-terminal tau antibody. (C) Reverse transcription (RT)-PCR to confirm Tau35 expression. Primers recognizing mouse tau (exons 7 and 13) generate a 612 bp band, corresponding to endogenous mouse tau in wild-type (WT) and Tau35 mice. Primers recognizing tau exon 9 and haemagglutinin generate a 558 bp band only in Tau35 mice. (D) RT-PCR of endogenous mouse and total tau in wild-type and Tau35 mice. PCR products amplified using primers for mouse tau (exons 7 and 13, 612 bp) or total tau (exons 9 and 13, 348 bp) in wild-type and Tau35 mice, standardized to glyceraldehyde 3-phosphate dehydrogenase (GAPDH). Results are shown as mean ± standard error of the mean (SEM). (n = 6 mice for each genotype). (E) Sagittal sections show widespread haemagglutinin labelling in Tau35 mouse brain (upper panels, scale bar = 2 mm). Higher magnifications of the hippocampal CA1 region show strongly haemagglutinin-positive pyramidal neurons in Tau35 mice (lower panels, scale bar = 200 μm). Western blots of frontal region and hippocampus/associated cortex (HC) show haemagglutinin protein expression only in Tau35 mice.
Figure 2
Figure 2
Progressive neuromuscular impairment and reduced survival of Tau35 mice. (A) Limb clasping is apparent in Tau35 mice from 2 months of age (image shows 8 months), with all Tau35 animals affected by 18 months (n = 40 mice). Clasping is not observed in wild-type (WT) mice at any age examined. (B) Spine curvature is apparent in Tau35 but not wild-type mice, at 14 months of age. A progressive reduction in the kyphotic index in Tau35 mice after 4 months of age indicates increasing spine curvature. Values shown are mean ± SEM, n = 8 mice for each genotype. (C) Kaplan-Meier survival plots reveal a median lifespan of 717 days for Tau35 mice, compared to 788 days for wild-type mice (P < 0.05, log-rank test, pairwise multiple comparison and Bonferroni correction), n = 10 mice for each genotype. (D) Tau35 mice have a significantly reduced latency to fall from an accelerating rotarod at all ages tested (4–16 months). Values shown are mean ± SEM, n = 8 mice for each genotype. (E) The grip strength of Tau35 mice declines steadily with age. Values shown are mean ± SEM, n = 8 for each genotype. (F) Haematoxylin and eosin staining of quadriceps and latissimus muscle sections from wild-type and Tau35 mice at 8 and 16 months. Muscle fibres from Tau35 mice show internal nuclei at 8 and 16 months (arrows). Scale bar = 60 μm. Graphs show increased internal nuclei (8 and 16 months) and altered distribution of muscle fibre diameter (minimal Feret’s diameter, 16 months) in Tau35 mice. Values shown are mean ± SEM, n = 3 mice for each genotype. *P < 0.05, **P < 0.01, ***P < 0.001.
Figure 3
Figure 3
Tau35 mice exhibit reduced hippocampal-dependent spatial learning ability in the Morris water maze. (A) Visible platform (VP) training in the Morris water maze was followed by 4 days of hidden platform training. Escape latencies were tracked for independent cohorts of Tau35 and wild-type (WT) mice at 4, 6, 8, 10 and 12 months of age. At 8, 10 and 12 months of age, Tau35 mice exhibit longer escape latency on the fourth day of testing compared to wild-type mice. The distance swam by Tau35 mice searching for the hidden platform is significantly increased compared to wild-type mice at 8, 10 and 12 months of age. (B) During the probe trial (1 min), the percentage occupancy of the target quadrant is significantly reduced for Tau35 mice compared to wild-type mice at 8 months. Tau35 and wild-type mice exhibit equivalent swim speeds during the probe trial indicating no apparent impairment in swimming ability at the ages tested. Values shown in A and B are mean ± SEM, n = 8 mice for each genotype. *P < 0.05, **P < 0.01, ***P < 0.001.
Figure 4
Figure 4
Tau pathology in Tau35 mouse brain. CA1 hippocampal sections of Tau35 mice at 2, 8, and 14-16 months of age, and wild-type (WT) mice aged 16 months (right), labelled with tau antibodies, PHF1, TOC1, MC1, AT8 and TP007, and counterstained with haematoxylin. PHF1 revealed tau-positive labelling at 2 months. Tau-positive labelling of inclusions in Tau35 brain, including dystrophic neurites and neuropil threads, was apparent with antibodies PHF1, TOC1, MC1, AT8 and TP007 at 14-16 months (insets). PHF1 revealed increased cytoplasmic labelling at 2 months and tau-containing inclusions from 8 months, which were more abundant at 14–16 months. Wild-type mouse hippocampal sections showed no labelling of inclusions with these antibodies at 14–16 months of age (right). n = 3 mice for each genotype, scale bar = 200 µm.
Figure 5
Figure 5
Tau phosphorylation changes in Tau35 mouse hippocampus. Western blots of (A) PHF1, total tau, and β-actin, (B) TG3 and total tau, and (C) AT270 and total tau, reveal a significant increase in tau phosphorylation in Tau35 mice at each phosphoepitope, compared to wild-type (WT) mice, whereas the total amount of tau relative to actin is equivalent in both genotypes. (D) Western blots of Tau1 and total tau, show a reduction in tau phosphorylation in Tau35 mice. Values shown are mean ± SEM, n = 6 mice for each genotype. *P < 0.05, ***P < 0.001.
Figure 6
Figure 6
Biochemical changes in Tau35 mouse hippocampus. Western blots show (A) phosphorylated (inactive) glycogen synthase kinase-3β (pGSK3β) relative to total (T-) GSK3β, is significantly decreased in Tau35 mice, indicating increased GSK3β activity. The total amounts of GSK3α and GSK3β, and of pGSK3α, are unchanged in Tau35 mice. (B) Western blot shows the amounts of LC3-I and LC3-II, relative to actin, are increased in Tau35 mice. (C) p62 is significantly increased, relative to actin, in Tau35 mice. (D) Mature (active) cathepsin D is reduced in Tau35 mice, whilst the amount of pro-cathepsin D is unchanged. (E) Western blots show a small but significant decrease in acetylated α-tubulin in Tau35 mice. The total amount of tubulin relative to actin is equivalent in both genotypes. (F) The amount of synapsin 1 is reduced relative to tubulin, whereas synaptophysin is unchanged in Tau35 mice (G). Values shown are mean ± SEM, n = 6 mice for each genotype. *P < 0.05, **P < 0.01, ***P < 0.001.
Figure 7
Figure 7
Phenylbutyrate rescues disease-related changes in Tau35 mice. (A) Morris water maze (10 months) testing of 4-phenylbutyrate (PBA, dotted lines) and vehicle-treated (solid lines) Tau35 (circles) and wild-type (WT, squares) mice. (B) Tau35 mice treated with PBA show improved learning in the probe trial at 10 months of age. (C) Grip strength of Tau35 (10 months) is restored by PBA treatment. Values shown in A–C are mean ± SEM, n = 8 mice for each group. Western blots show that Tau35 mice treated with PBA have (D) reduced phosphorylated tau (PHF1), relative to total tau, whereas the amounts of LC3-I and LC3-II are unchanged, relative to actin (E). PBA significantly reduced p62 in Tau35 mice compared to vehicle-treated animals (F). The amount of mature cathepsin D, but not pro-cathepsin D, is rescued by PBA treatment of Tau35 mice (G). Acetylated α-tubulin (H) and synapsin 1 (I) are both increased in Tau35 mice following PBA treatment, whereas the amount of synaptophysin is unchanged (J). Values shown in D–J are mean ± SEM, n = 5 mice for each group. (K) Haematoxylin and eosin staining of quadriceps muscle sections from Tau35 mice treated with vehicle (left) or PBA (right). Muscle fibres from vehicle-treated Tau35 mice show internal nuclei at 10 months (left, arrows). Scale bar = 60 μm. Graph shows reduced internal nuclei in Tau35 mice treated with PBA. Values shown are mean ± SEM, n = 3 mice for each group. *P < 0.05, **P < 0.01, ***P < 0.001.

References

    1. Arai T, Ikeda K, Akiyama H, Nonaka T, Hasegawa M, Ishiguro K, et al. Identification of amino-terminally cleaved tau fragments that distinguish progressive supranuclear palsy from corticobasal degeneration. Ann Neurol 2004; 55: 72–9.
    1. Azoulay-Alfaguter I, Elya R, Avrahami L, Katz A, Eldar-Finkelman H. Combined regulation of mTORC1 and lysosomal acidification by GSK-3 suppresses autophagy and contributes to cancer cell growth. Oncogene 2015; 34: 4613–23.
    1. Banreti A, Sass M, Graba Y. The emerging role of acetylation in the regulation of autophagy. Autophagy 2013; 9: 819–29.
    1. Bendiske J, Bahr BA. Lysosomal activation is a compensatory response against protein accumulation and associated synaptopathogenesis–an approach for slowing Alzheimer disease? J Neuropathol Exp Neurol 2003; 62: 451–63.
    1. Bhat R, Xue Y, Berg S, Hellberg S, Ormo M, Nilsson Y, et al. Structural insights and biological effects of glycogen synthase kinase 3-specific inhibitor AR-A014418. J Biol Chem 2003; 278: 45937–45.
    1. Bjorkoy G, Lamark T, Pankiv S, Overvatn A, Brech A, Johansen T. Monitoring autophagic degradation of p62/SQSTM1. Methods Enzymol 2009; 452: 181–97.
    1. Boland B, Kumar A, Lee S, Platt FM, Wegiel J, Yu WH, et al. Autophagy induction and autophagosome clearance in neurons: relationship to autophagic pathology in Alzheimer's disease. J Neurosci 2008; 28: 6926–37.
    1. Bronson SK, Plaehn EG, Kluckman KD, Hagaman JR, Maeda N, Smithies O. Single-copy transgenic mice with chosen-site integration. Proc Natl Acad Sci USA 1996; 93: 9067–72.
    1. Bruns MB, Josephs KA. Neuropsychiatry of corticobasal degeneration and progressive supranuclear palsy. Int Rev Psychiatry 2013; 25: 197–209.
    1. Burrell JR, Hodges JR, Rowe JB. Cognition in corticobasal syndrome and progressive supranuclear palsy: a review. Mov Disord 2014; 29: 684–93.
    1. Cesca F, Baldelli P, Valtorta F, Benfenati F. The synapsins: key actors of synapse function and plasticity. Prog Neurobiol 2010; 91: 313–48.
    1. Cho JA, Zhang X, Miller GM, Lencer WI, Nery FC. 4-phenylbutyrate attenuates the ER stress response and cyclic AMP accumulation in DYT1 dystonia cell models. PLoS One 2014; 9: e110086.
    1. Chung CW, Song YH, Kim IK, Yoon WJ, Ryu BR, Jo DG, et al. Proapoptotic effects of tau cleavage product generated by caspase-3. Neurobiol Dis 2001; 8: 162–72.
    1. Ciechanover A, Kwon YT. Degradation of misfolded proteins in neurodegenerative diseases: therapeutic targets and strategies. Exp Mol Med 2015; 47: e147.
    1. Cohen TJ, Guo JL, Hurtado DE, Kwong LK, Mills IP, Trojanowski JQ, et al. The acetylation of tau inhibits its function and promotes pathological tau aggregation. Nat Commun 2011; 2: 252.
    1. Dawson HN, Cantillana V, Chen L, Vitek MP. The tau N279K exon 10 splicing mutation recapitulates frontotemporal dementia and parkinsonism linked to chromosome 17 tauopathy in a mouse model. J Neurosci 2007; 27: 9155–68.
    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.
    1. Denk F, Wade-Martins R. Knock-out and transgenic mouse models of tauopathies. Neurobiol Aging 2009; 30: 1–13.
    1. Ebneth A, Godemann R, Stamer K, Illenberger S, Trinczek B, Mandelkow E. Overexpression of tau protein inhibits kinesin-dependent trafficking of vesicles, mitochondria, and endoplasmic reticulum: implications for Alzheimer's disease. J Cell Biol 1998; 143: 777–94.
    1. Fernandez-Nogales M, Cabrera JR, Santos-Galindo M, Hoozemans JJ, Ferrer I, Rozemuller AJ, et al. Huntington's disease is a four-repeat tauopathy with tau nuclear rods. Nat Med 2014; 20: 881–5.
    1. Goedert M, Spillantini MG. Pathogenesis of the tauopathies. J Mol Neurosci 2011; 45: 425–31.
    1. Goetzl EJ, Dolezalova H, Kong Y, Hu YL, Jaffe RB, Kalli KR, et al. Distinctive expression and functions of the type 4 endothelial differentiation gene-encoded G protein-coupled receptor for lysophosphatidic acid in ovarian cancer. Cancer Res 1999; 59: 5370–5.
    1. Gotz J, Deters N, Doldissen A, Bokhari L, Ke Y, Wiesner A, et al. A decade of tau transgenic animal models and beyond. Brain Pathol 2007; 17: 91–103.
    1. Hanger DP, Hughes K, Woodgett JR, Brion JP, Anderton BH. Glycogen synthase kinase-3 induces Alzheimer's disease-like phosphorylation of tau: generation of paired helical filament epitopes and neuronal localisation of the kinase. Neurosci Lett 1992; 147: 58–62.
    1. Hempen B, Brion JP. Reduction of acetylated à-tubulin immunoreactivity in neurofibrillary tangle-bearing neurons in Alzheimer's disease. J Neuropathol Exp Neurol 1996; 55: 964–72.
    1. Hockly E, Richon VM, Woodman B, Smith DL, Zhou X, Rosa E, et al. Suberoylanilide hydroxamic acid, a histone deacetylase inhibitor, ameliorates motor deficits in a mouse model of Huntington's disease. Proc Natl Acad Sci USA 2003; 100: 2041–6.
    1. Hyman BT, Augustinack JC, Ingelsson M. Transcriptional and conformational changes of the tau molecule in Alzheimer's disease. Biochim Biophys Acta 2005; 1739: 150–7.
    1. Iannitti T, Palmieri B. Clinical and experimental applications of sodium phenylbutyrate. Drugs R D 2011; 11: 227–49.
    1. Inoue K, Rispoli J, Kaphzan H, Klann E, Chen EI, Kim J, et al. Macroautophagy deficiency mediates age-dependent neurodegeneration through a phospho-tau pathway. Mol Neurodegener 2012; 7: 48.
    1. Kenessey A, Nacharaju P, Ko LW, Yen SH. Degradation of tau by lysosomal enzyme cathepsin D: implication for Alzheimer neurofibrillary degeneration. J Neurochem 1997; 69: 2026–38.
    1. Komatsu M, Waguri S, Koike M, Sou YS, Ueno T, Hara T, et al. Homeostatic levels of p62 control cytoplasmic inclusion body formation in autophagy-deficient mice. Cell 2007; 131: 1149–63.
    1. Korolchuk VI, Mansilla A, Menzies FM, Rubinsztein DC. Autophagy inhibition compromises degradation of ubiquitin-proteasome pathway substrates. Mol Cell 2009; 33: 517–27.
    1. Kuusisto E, Salminen A, Alafuzoff I. Early accumulation of p62 in neurofibrillary tangles in Alzheimer's disease: possible role in tangle formation. Neuropathol Appl Neurobiol 2002; 28: 228–37.
    1. Kwon SE, Chapman ER. Synaptophysin regulates the kinetics of synaptic vesicle endocytosis in central neurons. Neuron 2011; 70: 847–54.
    1. Lalonde R, Fukuchi K, Strazielle C. Neurologic and motor dysfunctions in APP transgenic mice. Rev Neurosci 2012; 23: 363–79.
    1. Lalonde R, Strazielle C. Brain regions and genes affecting limb-clasping responses. Brain Res Rev 2011; 67: 252–9.
    1. Laws N, Cornford-Nairn RA, Irwin N, Johnsen R, Fletcher S, Wilton SD, et al. Long-term administration of antisense oligonucleotides into the paraspinal muscles of mdx mice reduces kyphosis. J Appl Physiol 2008; 105: 662–8.
    1. Laws N, Hoey A. Progression of kyphosis in mdx mice. J Appl Physiol 2004; 97: 1970–7.
    1. Leclerc S, Garnier M, Hoessel R, Marko D, Bibb JA, Snyder GL, et al. Indirubins inhibit glycogen synthase kinase-3 beta and CDK5/p25, two protein kinases involved in abnormal tau phosphorylation in Alzheimer's disease. A property common to most cyclin-dependent kinase inhibitors? J Biol Chem 2001; 276: 251–60.
    1. Lee VM, Goedert M, Trojanowski JQ. Neurodegenerative tauopathies. Annu Rev Neurosci 2001; 24: 1121–59.
    1. Lein ES, Hawrylycz MJ, Ao N, Ayres M, Bensinger A, Bernard A, et al. Genome-wide atlas of gene expression in the adult mouse brain. Nature 2007; 445: 168–76. Available from:
    1. Lewis J, McGowan E, Rockwood J, Melrose H, Nacharaju P, Van Slegtenhorst M, et al. Neurofibrillary tangles, amyotrophy and progressive motor disturbance in mice expressing mutant (P301L) tau protein. Nat Genet 2000; 25: 402–5.
    1. Litvan I, Kong M. Rate of decline in progressive supranuclear palsy. Mov Disord 2014; 29: 463–8.
    1. Lovestone S, Reynolds CH, Latimer D, Davis DR, Anderton BH, Gallo JM, et al. Alzheimer's disease-like phosphorylation of the microtubule- associated protein tau by glycogen synthase kinase-3 in transfected mammalian cells. Curr Biol 1994; 4: 1077–86.
    1. Maeda S, Djukic B, Taneja P, Yu GQ, Lo I, Davis A, et al. Expression of A152T human tau causes age-dependent neuronal dysfunction and loss in transgenic mice. EMBO Rep 2016; 17: 530–51.
    1. Mandelkow EM, Drewes G, Biernat J, Gustke N, Van Lint J, Vandenheede JR, et al. Glycogen synthase kinase-3 and the Alzheimer-like state of microtubule-associated protein tau. FEBS Lett 1992; 314: 315–21.
    1. Marchand B, Arsenault D, Raymond-Fleury A, Boisvert FM, Boucher MJ. Glycogen synthase kinase-3 (GSK3) inhibition induces prosurvival autophagic signals in human pancreatic cancer cells. J Biol Chem 2015; 290: 5592–605.
    1. Melis V, Zabke C, Stamer K, Magbagbeolu M, Schwab K, Marschall P, et al. Different pathways of molecular pathophysiology underlie cognitive and motor tauopathy phenotypes in transgenic models for Alzheimer's disease and frontotemporal lobar degeneration. Cell Mol Life Sci 2015; 72: 2199–222.
    1. Miller N, Feng Z, Edens BM, Yang B, Shi H, Sze CC, et al. Non-aggregating tau phosphorylation by cyclin-dependent kinase 5 contributes to motor neuron degeneration in spinal muscular atrophy. J Neurosci 2015; 35: 6038–50.
    1. Mimori S, Okuma Y, Kaneko M, Kawada K, Hosoi T, Ozawa K, et al. Protective effects of 4-phenylbutyrate derivatives on the neuronal cell death and endoplasmic reticulum stress. Biol Pharm Bull 2012; 35: 84–90.
    1. Mizushima N, Yoshimori T. How to interpret LC3 immunoblotting. Autophagy 2007; 3: 542–5.
    1. Neary D, Snowden JS, Gustafson L, Passant U, Stuss D, Black S, et al. Frontotemporal lobar degeneration: a consensus on clinical diagnostic criteria. Neurology 1998; 51: 1546–54.
    1. Nixon RA. Autophagy, amyloidogenesis and Alzheimer disease. J Cell Sci 2007; 120(Pt 23): 4081–91.
    1. Nixon RA, Yang DS. Autophagy failure in Alzheimer's disease–locating the primary defect. Neurobiol Dis 2011; 43: 38–45.
    1. Noble W, Hanger DP, Gallo JM. Transgenic mouse models of tauopathy in drug discovery. CNS Neurol Disord Drug Targets 2010; 9: 403–28.
    1. Parr C, Carzaniga R, Gentleman SM, Van Leuven F, Walter J, Sastre M. Glycogen synthase kinase 3 inhibition promotes lysosomal biogenesis and autophagic degradation of the amyloid-beta precursor protein. Mol Cell Biol 2012; 32: 4410–8.
    1. Piras A, Collin L, Gruninger F, Graff C, Ronnback A. Autophagic and lysosomal defects in human tauopathies: analysis of post-mortem brain from patients with familial Alzheimer disease, corticobasal degeneration and progressive supranuclear palsy. Acta Neuropathol Commun 2016; 4: 22.
    1. Polito VA, Li H, Martini-Stoica H, Wang B, Yang L, Xu Y, et al. Selective clearance of aberrant tau proteins and rescue of neurotoxicity by transcription factor EB. EMBO Mol Med 2014; 6: 1142–60.
    1. Reed NA, Cai D, Blasius TL, Jih GT, Meyhofer E, Gaertig J, et al. Microtubule acetylation promotes kinesin-1 binding and transport. Curr Biol 2006; 16: 2166–72.
    1. Richter-Landsberg C, Leyk J. Inclusion body formation, macroautophagy, and the role of HDAC6 in neurodegeneration. Acta Neuropathol 2013; 126: 793–807.
    1. Ricobaraza A, Cuadrado-Tejedor M, Marco S, Perez-Otano I, Garcia-Osta A. Phenylbutyrate rescues dendritic spine loss associated with memory deficits in a mouse model of Alzheimer disease. Hippocampus 2012; 22: 1040–50.
    1. Ricobaraza A, Cuadrado-Tejedor M, Perez-Mediavilla A, Frechilla D, Del Rio J, Garcia-Osta A. Phenylbutyrate ameliorates cognitive deficit and reduces tau pathology in an Alzheimer's disease mouse model. Neuropsychopharmacology 2009; 34: 1721–32.
    1. Rosenmann H, Grigoriadis N, Eldar-Levy H, Avital A, Rozenstein L, Touloumi O, et al. A novel transgenic mouse expressing double mutant tau driven by its natural promoter exhibits tauopathy characteristics. Exp Neurol 2008; 212: 71–84.
    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.
    1. Schaeffer V, Lavenir I, Ozcelik S, Tolnay M, Winkler DT, Goedert M. Stimulation of autophagy reduces neurodegeneration in a mouse model of human tauopathy. Brain 2012; 135(Pt 7): 2169–77.
    1. Schindowski K, Bretteville A, Leroy K, Begard S, Brion JP, Hamdane M, et al. Alzheimer's disease-like tau neuropathology leads to memory deficits and loss of functional synapses in a novel mutated tau transgenic mouse without any motor deficits. Am J Pathol 2006; 169: 599–616.
    1. Terwel D, Lasrado R, Snauwaert J, Vandeweert E, Van HC, Borghgraef P, et al. Changed conformation of mutant Tau-P301L underlies the moribund tauopathy, absent in progressive, nonlethal axonopathy of Tau-4R/2N transgenic mice. J Biol Chem 2005; 280: 3963–73.
    1. Vianello S, Bouyon S, Benoit E, Sebrie C, Boerio D, Herbin M, et al. Arginine butyrate per os protects mdx mice against cardiomyopathy, kyphosis and changes in axonal excitability. Neurobiol Dis 2014; 71: 325–33.
    1. Vilchez D, Saez I, Dillin A. The role of protein clearance mechanisms in organismal ageing and age–related diseases. Nat Commun 2014; 5: 5659.
    1. Wiley JC, Meabon JS, Frankowski H, Smith EA, Schecterson LC, Bothwell M, et al. Phenylbutyric acid rescues endoplasmic reticulum stress-induced suppression of APP proteolysis and prevents apoptosis in neuronal cells. PLoS One 2010; 5: e9135.
    1. Wischik CM, Novak M, Thogersen HC, Edwards PC, Runswick MJ, Jakes R, et al. Isolation of a fragment of tau derived from the core of the paired helical filament of Alzheimer disease. Proc Natl Acad Sci USA 1988; 85: 4506–10.
    1. Wray S, Saxton M, Anderton B, Hanger D. Direct analysis of tau from PSP brain identifies new phosphorylation sites and a major fragment of N-terminally cleaved tau containing four microtubule-binding repeats. J Neurochem 2008; 105: 2343–52.
    1. Wu LS, Cheng WC, Shen CK. Targeted depletion of TDP-43 expression in the spinal cord motor neurons leads to the development of amyotrophic lateral sclerosis-like phenotypes in mice. J Biol Chem 2012; 287: 27335–44.
    1. Xie R, Nguyen S, McKeehan WL, Liu L. Acetylated microtubules are required for fusion of autophagosomes with lysosomes. BMC Cell Biol 2010; 11: 89.
    1. Yan J, Seibenhener ML, Calderilla-Barbosa L, Diaz-Meco MT, Moscat J, Jiang J, et al. SQSTM1/p62 interacts with HDAC6 and regulates deacetylase activity. PLoS One 2013; 8: e76016.
    1. Yang M, Crawley JN. Simple behavioral assessment of mouse olfaction. Curr Protoc Neurosci 2009; Chapter 8: Unit 8.24.
    1. Yoon SY, Kim DH. Alzheimer's disease genes and autophagy. Brain Res 2016. Advance Access published on March 23, 2016.
    1. Yoshiyama Y, Higuchi M, Zhang B, Huang SM, Iwata N, Saido TC, et al. Synapse loss and microglial activation precede tangles in a P301S tauopathy mouse model. Neuron 2007; 53: 337–51.
    1. Zhang Z, Song M, Liu X, Kang SS, Kwon IS, Duong DM, et al. Cleavage of tau by asparagine endopeptidase mediates the neurofibrillary pathology in Alzheimer's disease. Nat Med 2014; 20: 1254–62.
    1. Zilka N, Filipcik P, Koson P, Fialova L, Skrabana R, Zilkova M, et al. Truncated tau from sporadic Alzheimer's disease suffices to drive neurofibrillary degeneration in vivo. FEBS Lett 2006; 580: 3582–8.
    1. Zilka N, Korenova M, Novak M. Misfolded tau protein and disease modifying pathways in transgenic rodent models of human tauopathies. Acta Neuropathol 2009; 118: 71–86.

Source: PubMed

3
Předplatit