Progress in the treatment of Parkinson-Plus syndromes
Tài liệu tham khảo
Litvan, 2005, What is an atypical parkinsonian disorder?, 1
Wenning, 2011, Milestones in atypical and secondary Parkinsonisms, Mov. Disord., 26, 1083, 10.1002/mds.23713
McFarland, 2016, Diagnostic approach to atypical parkinsonian syndromes, Continuum (Minneap Minn), 22, 1117
Giagkou, 2018
Gilman, 2008, Second consensus statement on the diagnosis of multiple system atrophy, Neurology, 71, 670, 10.1212/01.wnl.0000324625.00404.15
Fanciulli, 2014, Clinical presentation, 97
McKeith, 2017, Diagnosis and management of dementia with Lewy bodies: fourth consensus report of the DLB Consortium, Neurology, 89, 88, 10.1212/WNL.0000000000004058
Fujishiro, 2008, Validation of the neuropathologic criteria of the third consortium for dementia with Lewy bodies for prospectively diagnosed cases, J. Neuropathol. Exp. Neurol., 67, 649, 10.1097/NEN.0b013e31817d7a1d
Asi, 2014, Alpha-synuclein mRNA expression in oligodendrocytes in MSA, Glia, 62, 964, 10.1002/glia.22653
Reyes, 2014, Alpha-synuclein transfers from neurons to oligodendrocytes, Glia, 62, 387, 10.1002/glia.22611
Bartels, 2011, alpha-Synuclein occurs physiologically as a helically folded tetramer that resists aggregation, Nature, 477, 107, 10.1038/nature10324
Burre, 2018, Cell biology and pathophysiology of alpha-synuclein, Cold Spring Harb. Perspect. Med., 8, 10.1101/cshperspect.a024091
Ueda, 1993, Molecular cloning of cDNA encoding an unrecognized component of amyloid in Alzheimer disease, Proc. Natl. Acad. Sci. U. S. A., 90, 11282, 10.1073/pnas.90.23.11282
Burre, 2015, The synaptic function of alpha-synuclein, J. Parkinson's Dis., 5, 699, 10.3233/JPD-150642
Hishikawa, 2003, Clinical and neuropathological correlates of Lewy body disease, Acta Neuropathol., 105, 341, 10.1007/s00401-002-0651-4
Ozawa, 2004, The spectrum of pathological involvement of the striatonigral and olivopontocerebellar systems in multiple system atrophy: clinicopathological correlations, Brain, 127, 2657, 10.1093/brain/awh303
Jellinger, 2009, A critical evaluation of current staging of alpha-synuclein pathology in Lewy body disorders, Biochim. Biophys. Acta, 1792, 730, 10.1016/j.bbadis.2008.07.006
Chui, 1994, Decreased beta-amyloid and increased abnormal Tau deposition in the brain of aged patients with leprosy, Am. J. Pathol., 145, 771
Tomiyama, 1996, Inhibition of amyloid beta protein aggregation and neurotoxicity by rifampicin. Its possible function as a hydroxyl radical scavenger, J. Biol. Chem., 271, 6839, 10.1074/jbc.271.12.6839
Li, 2004, Rifampicin inhibits alpha-synuclein fibrillation and disaggregates fibrils, Chem. Biol., 11, 1513, 10.1016/j.chembiol.2004.08.025
Ubhi, 2008, Rifampicin reduces alpha-synuclein in a transgenic mouse model of multiple system atrophy, Neuroreport, 19, 1271, 10.1097/WNR.0b013e32830b3661
Low, 2014, Efficacy and safety of rifampicin for multiple system atrophy: a randomised, double-blind, placebo-controlled trial, Lancet Neurol., 13, 268, 10.1016/S1474-4422(13)70301-6
Bieschke, 2010, EGCG remodels mature alpha-synuclein and amyloid-beta fibrils and reduces cellular toxicity, Proc. Natl. Acad. Sci. U. S. A., 107, 7710, 10.1073/pnas.0910723107
Caruana, 2011, Inhibition and disaggregation of alpha-synuclein oligomers by natural polyphenolic compounds, FEBS Lett., 585, 1113, 10.1016/j.febslet.2011.03.046
Levin, 2016, The PROMESA-protocol: progression rate of multiple system atrophy under EGCG supplementation as anti-aggregation-approach, J. Neural. Transm. (Vienna), 123, 439, 10.1007/s00702-016-1507-8
Schuberth, 2017, PROMESA: progression Rate of MSA under EGCG Supplementation as anti-Aggregation-Approach – evaluation of serious adverse events [abstract]
Schwarz, 2012, Involvement of macroautophagy in multiple system atrophy and protein aggregate formation in oligodendrocytes, J. Mol. Neurosci., 47, 256, 10.1007/s12031-012-9733-5
Webb, 2003, Alpha-Synuclein is degraded by both autophagy and the proteasome, J. Biol. Chem., 278, 25009, 10.1074/jbc.M300227200
Cuervo, 2004, Impaired degradation of mutant alpha-synuclein by chaperone-mediated autophagy, Science, 305, 1292, 10.1126/science.1101738
Klucken, 2012, Alpha-synuclein aggregation involves a bafilomycin A 1-sensitive autophagy pathway, Autophagy, 8, 754, 10.4161/auto.19371
Lee, 2013, Autophagic failure promotes the exocytosis and intercellular transfer of alpha-synuclein, Exp. Mol. Med., 45, e22, 10.1038/emm.2013.45
Sarkar, 2005, Lithium induces autophagy by inhibiting inositol monophosphatase, J. Cell Biol., 170, 1101, 10.1083/jcb.200504035
Verstraete, 2012, Lithium lacks effect on survival in amyotrophic lateral sclerosis: a phase IIb randomised sequential trial, J. Neurol. Neurosurg. Psychiatry, 83, 557, 10.1136/jnnp-2011-302021
Matsunaga, 2015, Lithium as a treatment for alzheimer's disease: a systematic review and meta-analysis, J Alzheimers Dis, 48, 403, 10.3233/JAD-150437
Sacca, 2013, A randomized clinical trial of lithium in multiple system atrophy, J. Neurol., 260, 458, 10.1007/s00415-012-6655-7
Bae, 2012, Antibody-aided clearance of extracellular alpha-synuclein prevents cell-to-cell aggregate transmission, J. Neurosci., 32, 13454, 10.1523/JNEUROSCI.1292-12.2012
Masliah, 2011, Passive immunization reduces behavioral and neuropathological deficits in an alpha-synuclein transgenic model of Lewy body disease, PloS One, 6, 10.1371/journal.pone.0019338
Mandler, 2014, Next-generation active immunization approach for synucleinopathies: implications for Parkinson's disease clinical trials, Acta Neuropathol., 127, 861, 10.1007/s00401-014-1256-4
Games, 2014, Reducing C-terminal-truncated alpha-synuclein by immunotherapy attenuates neurodegeneration and propagation in Parkinson's disease-like models, J. Neurosci., 34, 9441, 10.1523/JNEUROSCI.5314-13.2014
Peelaerts, 2015, alpha-Synuclein strains cause distinct synucleinopathies after local and systemic administration, Nature, 522, 340, 10.1038/nature14547
Peelaerts, 2018, a-Synuclein strains and seeding in Parkinson's disease, incidental Lewy body disease, dementia with Lewy bodies and multiple system atrophy: similarities and differences, Cell Tissue Res., 373, 195, 10.1007/s00441-018-2839-5
Mandler, 2015, Active immunization against alpha-synuclein ameliorates the degenerative pathology and prevents demyelination in a model of multiple system atrophy, Mol. Neurodegener., 10, 10, 10.1186/s13024-015-0008-9
Affiris
Affiris, 2018
Gerhard, 2003, [11C](R)-PK11195 PET imaging of microglial activation in multiple system atrophy, Neurology, 61, 686, 10.1212/01.WNL.0000078192.95645.E6
Ishizawa, 2004, Microglial activation parallels system degeneration in multiple system atrophy, J. Neuropathol. Exp. Neurol., 63, 43, 10.1093/jnen/63.1.43
Tang, 2016, Differential roles of M1 and M2 microglia in neurodegenerative diseases, Mol. Neurobiol., 53, 1181, 10.1007/s12035-014-9070-5
Kaindlstorfer, 2015, Failure of neuroprotection despite microglial suppression by delayed-start myeloperoxidase inhibition in a model of advanced multiple system atrophy: clinical implications, Neurotox. Res., 28, 185, 10.1007/s12640-015-9547-7
Stefanova, 2012, Myeloperoxidase inhibition ameliorates multiple system atrophy-like degeneration in a transgenic mouse model, Neurotox. Res., 21, 393, 10.1007/s12640-011-9294-3
Dodel, 2010, Minocycline 1-year therapy in multiple-system-atrophy: effect on clinical symptoms and [(11)C] (R)-PK11195 PET (MEMSA-trial), Mov. Disord., 25, 97, 10.1002/mds.22732
Biohaven
Novak, 2012, Treatment of multiple system atrophy using intravenous immunoglobulin, BMC Neurol., 12, 131, 10.1186/1471-2377-12-131
Harvey, 1993, Growth hormone: neurocrine and neuroendocrine perspectives, Growth Regul., 3, 161
Aberg, 2000, Peripheral infusion of IGF-I selectively induces neurogenesis in the adult rat hippocampus, J. Neurosci., 20, 2896, 10.1523/JNEUROSCI.20-08-02896.2000
Ajo, 2003, Growth hormone action on proliferation and differentiation of cerebral cortical cells from fetal rat, Endocrinology, 144, 1086, 10.1210/en.2002-220667
Holmberg, 2007, Safety and tolerability of growth hormone therapy in multiple system atrophy: a double-blind, placebo-controlled study, Mov. Disord., 22, 1138, 10.1002/mds.21501
Caumont, 2006, Specific regulation of rat glial cell line-derived neurotrophic factor gene expression by riluzole in C6 glioma cells, J. Neurochem., 97, 128, 10.1111/j.1471-4159.2006.03711.x
Shortland, 2006, Riluzole promotes cell survival and neurite outgrowth in rat sensory neurones in vitro, Eur. J. Neurosci., 24, 3343, 10.1111/j.1460-9568.2006.05218.x
Diguet, 2005, Effects of riluzole on combined MPTP + 3-nitropropionic acid-induced mild to moderate striatonigral degeneration in mice, J Neural Transm (Vienna), 112, 613, 10.1007/s00702-004-0206-z
Scherfler, 2005, Riluzole improves motor deficits and attenuates loss of striatal neurons in a sequential double lesion rat model of striatonigral degeneration (Parkinson variant of multiple system atrophy), J Neural Transm (Vienna), 112, 1025, 10.1007/s00702-004-0245-5
Bensimon, 2009, Riluzole treatment, survival and diagnostic criteria in Parkinson plus disorders: the NNIPPS study, Brain, 132, 156, 10.1093/brain/awn291
Youdim, 2001, Rasagiline [N-propargyl-1R(+)-aminoindan], a selective and potent inhibitor of mitochondrial monoamine oxidase B, Br. J. Pharmacol., 132, 500, 10.1038/sj.bjp.0703826
Youdim, 2001, Molecular basis of neuroprotective activities of rasagiline and the anti-Alzheimer drug TV3326 [(N-propargyl-(3R)aminoindan-5-YL)-ethyl methyl carbamate], Cell. Mol. Neurobiol., 21, 555, 10.1023/A:1015131516649
Stefanova, 2008, Rasagiline is neuroprotective in a transgenic model of multiple system atrophy, Exp. Neurol., 210, 421, 10.1016/j.expneurol.2007.11.022
Poewe, 2015, Efficacy of rasagiline in patients with the parkinsonian variant of multiple system atrophy: a randomised, placebo-controlled trial, Lancet Neurol., 14, 145, 10.1016/S1474-4422(14)70288-1
Ubhi, 2012, Fluoxetine ameliorates behavioral and neuropathological deficits in a transgenic model mouse of alpha-synucleinopathy, Exp. Neurol., 234, 405, 10.1016/j.expneurol.2012.01.008
Valera, 2014, Antidepressants reduce neuroinflammatory responses and astroglial alpha-synuclein accumulation in a transgenic mouse model of multiple system atrophy, Glia, 62, 317, 10.1002/glia.22610
Stefanova, 2018, Translational therapies for multiple system atrophy: bottlenecks and future directions, Auton. Neurosci., 211, 7, 10.1016/j.autneu.2017.09.016
Lee, 2008, Autologous mesenchymal stem cell therapy delays the progression of neurological deficits in patients with multiple system atrophy, Clin. Pharmacol. Ther., 83, 723, 10.1038/sj.clpt.6100386
Lee, 2012, A randomized trial of mesenchymal stem cells in multiple system atrophy, Ann. Neurol., 72, 32, 10.1002/ana.23612
Singer, 2017, Intrathecal administration of autologous mesenchymal stem cells in multiple system atrophy – a phase I/II dose-escalation trial [abstract]
Singer, 2018, Intrathecal administration of autologous mesenchymal stem cells in multiple system atrophy results in growth factor spike [abstract]
Lee, 2011, Uric acid as a potential disease modifier in patients with multiple system atrophy, Mov. Disord., 26, 1533, 10.1002/mds.23556
Cao, 2013, Uric acid is associated with the prevalence but not disease progression of multiple system atrophy in Chinese population, J. Neurol., 260, 2511, 10.1007/s00415-013-7006-z
Cao, 2015, Association of serum uric acid level with cognitive function among patients with multiple system atrophy, J. Neurol. Sci., 359, 363, 10.1016/j.jns.2015.11.025
Chen, 2015, Contra-directional expression of serum homocysteine and uric acid as important biomarkers of multiple system atrophy severity: a cross-sectional study, Front. Cell. Neurosci., 9, 247, 10.3389/fnagi.2017.00247
Fukae, 2017, Serum uric acid level is linked to the disease progression rate in male patients with multiple system atrophy, Clin. Neurol. Neurosurg., 158, 15, 10.1016/j.clineuro.2017.04.002
Bassil, 2016, Reducing C-terminal truncation mitigates synucleinopathy and neurodegeneration in a transgenic model of multiple system atrophy, Proc. Natl. Acad. Sci. U. S. A., 113, 9593, 10.1073/pnas.1609291113
Fellner, 2016, Anle138b partly ameliorates motor deficits despite failure of neuroprotection in a model of advanced multiple system atrophy, Front. Neurosci., 10, 99, 10.3389/fnins.2016.00099
Wang, 2016, Caspase-1 causes truncation and aggregation of the Parkinson's disease-associated protein alpha-synuclein, Proc. Natl. Acad. Sci. U. S. A., 113, 9587, 10.1073/pnas.1610099113
Stefanova, 2017, Targeting alpha-synuclein pathology with the molecular tweezer CLR01 in multiple system atrophy, 58
Hoyer, 2004, Impact of the acidic C-terminal region comprising amino acids 109-140 on alpha-synuclein aggregation in vitro, Biochemistry, 43, 16233, 10.1021/bi048453u
Liu, 2005, A precipitating role for truncated alpha-synuclein and the proteasome in alpha-synuclein aggregation: implications for pathogenesis of Parkinson disease, J. Biol. Chem., 280, 22670, 10.1074/jbc.M501508200
Ulusoy, 2010, Co-expression of C-terminal truncated alpha-synuclein enhances full-length alpha-synuclein-induced pathology, Eur. J. Neurosci., 32, 409, 10.1111/j.1460-9568.2010.07284.x
Boxer, 2010
Valera, 2017, MicroRNA-101 modulates autophagy and oligodendroglial alpha-synuclein accumulation in multiple system atrophy, Front. Mol. Neurosci., 10, 329, 10.3389/fnmol.2017.00329
Hebron, 2013, Nilotinib reverses loss of dopamine neurons and improves motor behavior via autophagic degradation of alpha-synuclein in Parkinson's disease models, Hum. Mol. Genet., 22, 3315, 10.1093/hmg/ddt192
Pagan, 2016, Nilotinib effects in Parkinson's disease and dementia with Lewy bodies, J. Parkinson's Dis., 6, 503, 10.3233/JPD-160867
Ngoc, 2017, A case report of multiple system atrophy treated with an Abelson tyrosine kinase inhibitor, J. Neurol. Sci., 382, 53, 10.1016/j.jns.2017.09.013
Kawamoto, 2000, Glial cell line-derived neurotrophic factor-like immunoreactivity in the cerebella of normal subjects and patients with multiple system atrophy, Acta Neuropathol., 100, 131, 10.1007/s004010050004
Ubhi, 2010, Neurodegeneration in a transgenic mouse model of multiple system atrophy is associated with altered expression of oligodendroglial-derived neurotrophic factors, J. Neurosci., 30, 6236, 10.1523/JNEUROSCI.0567-10.2010
Sailer, 2016, A genome-wide association study in multiple system atrophy, Neurology, 87, 1591, 10.1212/WNL.0000000000003221
Kontopoulos, 2006, Alpha-synuclein acts in the nucleus to inhibit histone acetylation and promote neurotoxicity, Hum. Mol. Genet., 15, 3012, 10.1093/hmg/ddl243
Urdinguio, 2009, Epigenetic mechanisms in neurological diseases: genes, syndromes, and therapies, Lancet Neurol., 8, 1056, 10.1016/S1474-4422(09)70262-5
Sturm, 2014, Multiple system atrophy: genetic or epigenetic?, Exp Neurobiol, 23, 277, 10.5607/en.2014.23.4.277
Sturm, 2016, Neuroprotection by epigenetic modulation in a transgenic model of multiple system atrophy, Neurotherapeutics, 13, 871, 10.1007/s13311-016-0447-1
Jellinger, 2018, Are dementia with Lewy bodies and Parkinson's disease dementia the same disease?, BMC Med., 16, 34, 10.1186/s12916-018-1016-8
Games, 2013, Axonopathy in an alpha-synuclein transgenic model of Lewy body disease is associated with extensive accumulation of C-terminal-truncated alpha-synuclein, Am. J. Pathol., 182, 940, 10.1016/j.ajpath.2012.11.018
Fagerqvist, 2013, Monoclonal antibodies selective for alpha-synuclein oligomers/protofibrils recognize brain pathology in Lewy body disorders and alpha-synuclein transgenic mice with the disease-causing A30P mutation, J. Neurochem., 126, 131, 10.1111/jnc.12175
Fagerqvist, 2013, Off-pathway alpha-synuclein oligomers seem to alter alpha-synuclein turnover in a cell model but lack seeding capability in vivo, Amyloid, 20, 233, 10.3109/13506129.2013.835726
Lindstrom, 2014, Immunotherapy targeting alpha-synuclein protofibrils reduced pathology in (Thy-1)-h[A30P] alpha-synuclein mice, Neurobiol. Dis., 69, 134, 10.1016/j.nbd.2014.05.009
Ingelsson, 2016, Alpha-synuclein oligomers-neurotoxic molecules in Parkinson's disease and other Lewy body disorders, Front. Neurosci., 10, 408, 10.3389/fnins.2016.00408
Jankovic, 2018, Safety and tolerability of multiple ascending doses of PRX002/RG7935, an anti-alpha-synuclein monoclonal antibody, in patients with Parkinson disease: a randomized clinical trial, JAMA Neurol, 10.1001/jamaneurol.2018.1487
Brys, 2017, Randomized, double-blind, placebo-controlled, single ascending dose study of anti-alpha-synuclein antibody BIIB054 in healthy volunteers [abstract]
Alzforum
Colom-Cadena, 2013, Confluence of alpha-synuclein, tau, and beta-amyloid pathologies in dementia with Lewy bodies, J. Neuropathol. Exp. Neurol., 72, 1203, 10.1097/NEN.0000000000000018
Colom-Cadena, 2017, Regional overlap of pathologies in Lewy body disorders, J. Neuropathol. Exp. Neurol., 76, 216
Colom-Cadena, 2013, MAPT H1 haplotype is associated with enhanced alpha-synuclein deposition in dementia with Lewy bodies, Neurobiol. Aging, 34, 936, 10.1016/j.neurobiolaging.2012.06.015
Nath, 2001, The prevalence of progressive supranuclear palsy (Steele-Richardson-Olszewski syndrome) in the UK, Brain, 124, 1438, 10.1093/brain/124.7.1438
Golbe, 2008, The epidemiology of progressive supranuclear palsy, Handb. Clin. Neurol., 89, 457, 10.1016/S0072-9752(07)01242-0
Steele, 1964, Progressive supranuclear palsy. A heterogeneous degeneration involving the brain stem, basal ganglia and cerebellum with vertical gaze and pseudobulbar palsy, nuchal dystonia and dementia, Arch. Neurol., 10, 333, 10.1001/archneur.1964.00460160003001
Boxer, 2017, Advances in progressive supranuclear palsy: new diagnostic criteria, biomarkers, and therapeutic approaches, Lancet Neurol., 16, 552, 10.1016/S1474-4422(17)30157-6
Schoch, 2016, Increased 4R-tau induces pathological changes in a human-tau mouse model, Neuron, 90, 941, 10.1016/j.neuron.2016.04.042
Kovacs, 2017, Tauopathies, Handb Clin Neurol, 145, 355, 10.1016/B978-0-12-802395-2.00025-0
Rebeiz, 1967, Corticodentatonigral degeneration with neuronal achromasia: a progressive disorder of late adult life, Trans. Am. Neurol. Assoc., 92, 23
Armstrong, 2013, Criteria for the diagnosis of corticobasal degeneration, Neurology, 80, 496, 10.1212/WNL.0b013e31827f0fd1
Shoeibi, 2017, Therapeutic options for Progressive Supranuclear Palsy including investigational drugs, Expert Opin Orphan Drugs, 5, 575, 10.1080/21678707.2017.1335596
Coyle-Gilchrist, 2016, Prevalence, characteristics, and survival of frontotemporal lobar degeneration syndromes, Neurology, 86, 1736, 10.1212/WNL.0000000000002638
Arai, 2001, Intracellular processing of aggregated tau differs between corticobasal degeneration and progressive supranuclear palsy, Neuroreport, 12, 935, 10.1097/00001756-200104170-00014
Arai, 2004, Identification of amino-terminally cleaved tau fragments that distinguish progressive supranuclear palsy from corticobasal degeneration, Ann. Neurol., 55, 72, 10.1002/ana.10793
Wakabayashi, 2004, Pathological heterogeneity in progressive supranuclear palsy and corticobasal degeneration, Neuropathology, 24, 79, 10.1111/j.1440-1789.2003.00543.x
Shoeibi, 2018, Preclinical, phase I, and phase II investigational clinical trials for treatment of progressive supranuclear palsy, Expet Opin. Invest. Drugs, 27, 349, 10.1080/13543784.2018.1460356
Guo, 2017, Roles of tau protein in health and disease, Acta Neuropathol., 133, 665, 10.1007/s00401-017-1707-9
Forman, 2002, Signature tau neuropathology in gray and white matter of corticobasal degeneration, Am. J. Pathol., 160, 2045, 10.1016/S0002-9440(10)61154-6
Hanger, 2007, Novel phosphorylation sites in tau from Alzheimer brain support a role for casein kinase 1 in disease pathogenesis, J. Biol. Chem., 282, 23645, 10.1074/jbc.M703269200
Cohen, 2011, The acetylation of tau inhibits its function and promotes pathological tau aggregation, Nat. Commun., 2, 252, 10.1038/ncomms1255
Irwin, 2012, Acetylated tau, a novel pathological signature in Alzheimer's disease and other tauopathies, Brain, 135, 807, 10.1093/brain/aws013
Hastings, 2017, Inhibition of O-GlcNAcase leads to elevation of O-GlcNAc tau and reduction of tauopathy and cerebrospinal fluid tau in rTg4510 mice, Mol. Neurodegener., 12, 39, 10.1186/s13024-017-0181-0
Ferrer, 2002, Glycogen synthase kinase-3 is associated with neuronal and glial hyperphosphorylated tau deposits in Alzheimer's disease, Pick's disease, progressive supranuclear palsy and corticobasal degeneration, Acta Neuropathol., 104, 583, 10.1007/s00401-002-0587-8
Tolosa, 2014, A phase 2 trial of the GSK-3 inhibitor tideglusib in progressive supranuclear palsy, Mov. Disord., 29, 470, 10.1002/mds.25824
Leclair-Visonneau, 2016, Randomized placebo-controlled trial of sodium valproate in progressive supranuclear palsy, Clin. Neurol. Neurosurg., 146, 35, 10.1016/j.clineuro.2016.04.021
Min, 2010, Acetylation of tau inhibits its degradation and contributes to tauopathy, Neuron, 67, 953, 10.1016/j.neuron.2010.08.044
Yin, 1998, The anti-inflammatory agents aspirin and salicylate inhibit the activity of I(kappa)B kinase-beta, Nature, 396, 77, 10.1038/23948
Liu, 2004, O-GlcNAcylation regulates phosphorylation of tau: a mechanism involved in Alzheimer's disease, Proc. Natl. Acad. Sci. U. S. A., 101, 10804, 10.1073/pnas.0400348101
Smith, 2016, Early clinical results and preclinical validation of the O-GlcNAcase (OGA) inhibitor MK-8719 as a novel therapeutic for the treatment of tauopathies [abstract], 261
Ryan, 2018, Phase 1 study in healthy volunteers of the O-glcnacase inhibitor ASN120290 as a novel therapy for progressive supranuclear palsy and related tauopathies [abstract]
Clavaguera, 2009, Transmission and spreading of tauopathy in transgenic mouse brain, Nat. Cell Biol., 11, 909, 10.1038/ncb1901
Clavaguera, 2013, Brain homogenates from human tauopathies induce tau inclusions in mouse brain, Proc. Natl. Acad. Sci. U. S. A., 110, 9535, 10.1073/pnas.1301175110
Sanders, 2014, Distinct tau prion strains propagate in cells and mice and define different tauopathies, Neuron, 82, 1271, 10.1016/j.neuron.2014.04.047
Narasimhan, 2017, Pathological tau strains from human brains Recapitulate the diversity of tauopathies in nontransgenic mouse brain, J. Neurosci., 37, 11406, 10.1523/JNEUROSCI.1230-17.2017
Yanamandra, 2013, Anti-tau antibodies that block tau aggregate seeding in vitro markedly decrease pathology and improve cognition in vivo, Neuron, 80, 402, 10.1016/j.neuron.2013.07.046
Holmes, 2014, Prion-like properties of Tau protein: the importance of extracellular Tau as a therapeutic target, J. Biol. Chem., 289, 19855, 10.1074/jbc.R114.549295
Croft, 2018, Novel monoclonal antibodies targeting the microtubule-binding domain of human tau, PloS One, 13, 10.1371/journal.pone.0195211
Kanaan, 2015, Chapter 56 - tau protein: biology and pathobiology, 857
Santacruz, 2005, Tau suppression in a neurodegenerative mouse model improves memory function, Science, 309, 476, 10.1126/science.1113694
Gozes, 2004, The femtomolar-acting NAP interacts with microtubules: novel aspects of astrocyte protection, J Alzheimers Dis, 6, S37, 10.3233/JAD-2004-6S605
Shiryaev, 2009, NAP protects memory, increases soluble tau and reduces tau hyperphosphorylation in a tauopathy model, Neurobiol. Dis., 34, 381, 10.1016/j.nbd.2009.02.011
Boxer, 2014, Davunetide in patients with progressive supranuclear palsy: a randomised, double-blind, placebo-controlled phase 2/3 trial, Lancet Neurol., 13, 676, 10.1016/S1474-4422(14)70088-2
Boxer, 2017
Myeku, 2016, Tau-driven 26S proteasome impairment and cognitive dysfunction can be prevented early in disease by activating cAMP-PKA signaling, Nat. Med., 22, 46, 10.1038/nm.4011
Piras, 2016, 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, 4, 22, 10.1186/s40478-016-0292-9
Swerdlow, 2000, Mitochondrial dysfunction in cybrid lines expressing mitochondrial genes from patients with progressive supranuclear palsy, J. Neurochem., 75, 1681, 10.1046/j.1471-4159.2000.0751681.x
Stamelou, 2009, In vivo evidence for cerebral depletion in high-energy phosphates in progressive supranuclear palsy, J. Cerebr. Blood Flow Metabol., 29, 861, 10.1038/jcbfm.2009.2
Esteras, 2017, Mitochondrial hyperpolarization in iPSC-derived neurons from patients of FTDP-17 with 10+16 MAPT mutation leads to oxidative stress and neurodegeneration, Redox Biol, 12, 410, 10.1016/j.redox.2017.03.008
Stamelou, 2008, Short-term effects of coenzyme Q10 in progressive supranuclear palsy: a randomized, placebo-controlled trial, Mov. Disord., 23, 942, 10.1002/mds.22023
Apetauerova, 2016, CoQ10 in progressive supranuclear palsy: a randomized, placebo-controlled, double-blind trial, Neurol Neuroimmunol Neuroinflamm, 3, e266, 10.1212/NXI.0000000000000266
Piedrahita, 2010, Silencing of CDK5 reduces neurofibrillary tangles in transgenic alzheimer's mice, J. Neurosci., 30, 13966, 10.1523/JNEUROSCI.3637-10.2010
Georgievska, 2013, AZD1080, a novel GSK3 inhibitor, rescues synaptic plasticity deficits in rodent brain and exhibits peripheral target engagement in humans, J. Neurochem., 125, 446, 10.1111/jnc.12203
Bian, 2016, RNA interference silencing of glycogen synthase kinase 3beta inhibites tau phosphorylation in mice with alzheimer disease, Neurochem. Res., 41, 2470, 10.1007/s11064-016-1960-7
Gentry, 2016, Rho kinase inhibition as a therapeutic for progressive supranuclear palsy and corticobasal degeneration, J. Neurosci., 36, 1316, 10.1523/JNEUROSCI.2336-15.2016
Croft, 2017, Inhibition of glycogen synthase kinase-3 by BTA-EG4 reduces tau abnormalities in an organotypic brain slice culture model of Alzheimer's disease, Sci. Rep., 7, 7434, 10.1038/s41598-017-07906-1
Cisek, 2014, Structure and mechanism of action of tau aggregation inhibitors, Curr. Alzheimer Res., 11, 918, 10.2174/1567205011666141107150331
Seidler, 2018, Structure-based inhibitors of tau aggregation, Nat. Chem., 10, 170, 10.1038/nchem.2889
Kalbfuss, 2001, Correction of alternative splicing of tau in frontotemporal dementia and parkinsonism linked to chromosome 17, J. Biol. Chem., 276, 42986, 10.1074/jbc.M105113200
Sud, 2014, Antisense-mediated exon skipping decreases tau protein expression: a potential therapy for tauopathies, Mol. Ther. Nucleic Acids, 3, 10.1038/mtna.2014.30
Xu, 2014, Tau silencing by siRNA in the P301S mouse model of tauopathy, Curr. Gene Ther., 14, 343, 10.2174/156652321405140926160602
Harada, 1994, Altered microtubule organization in small-calibre axons of mice lacking tau protein, Nature, 369, 488, 10.1038/369488a0
Denk, 2009, Knock-out and transgenic mouse models of tauopathies, Neurobiol. Aging, 30, 1, 10.1016/j.neurobiolaging.2007.05.010
DeVos, 2013, Antisense reduction of tau in adult mice protects against seizures, J. Neurosci., 33, 12887, 10.1523/JNEUROSCI.2107-13.2013
Ishizawa, 2001, Microglial activation parallels system degeneration in progressive supranuclear palsy and corticobasal degeneration, J. Neuropathol. Exp. Neurol., 60, 647, 10.1093/jnen/60.6.647
Gerhard, 2006, In vivo imaging of microglial activation with [11C](R)-PK11195 PET in progressive supranuclear palsy, Mov. Disord., 21, 89, 10.1002/mds.20668
Broce, 2018, Immune-related genetic enrichment in frontotemporal dementia: an analysis of genome-wide association studies, PLoS Med., 15
Passamonti, 2018, [(11)C]PK11195 binding in Alzheimer disease and progressive supranuclear palsy, Neurology, 90, 10.1212/WNL.0000000000005610
Postuma, 2015, MDS clinical diagnostic criteria for Parkinson's disease, Mov. Disord., 30, 1591, 10.1002/mds.26424
Flabeau, 2010, Multiple system atrophy: current and future approaches to management, Ther Adv Neurol Disord, 3, 249, 10.1177/1756285610375328
Williams, 2005, Characteristics of two distinct clinical phenotypes in pathologically proven progressive supranuclear palsy: Richardson's syndrome and PSP-parkinsonism, Brain, 128, 1247, 10.1093/brain/awh488
Wenning, 1998, Natural history and survival of 14 patients with corticobasal degeneration confirmed at postmortem examination, J. Neurol. Neurosurg. Psychiatry, 64, 184, 10.1136/jnnp.64.2.184
Ling, 2010, Does corticobasal degeneration exist? A clinicopathological re-evaluation, Brain, 133, 2045, 10.1093/brain/awq123
Shehata, 2015, Corticobasal degeneration: clinical characteristics and multidisciplinary therapeutic approach in 26 patients, Neurol. Sci., 36, 1651, 10.1007/s10072-015-2226-x
Constantinescu, 2007, Levodopa responsiveness in disorders with parkinsonism: a review of the literature, Mov. Disord., 22, 2141, 10.1002/mds.21578
Kollensperger, 2010, Presentation, diagnosis, and management of multiple system atrophy in Europe: final analysis of the European multiple system atrophy registry, Mov. Disord., 25, 2604, 10.1002/mds.23192
Goldman, 2008, Effects of dopaminergic medications on psychosis and motor function in dementia with Lewy bodies, Mov. Disord., 23, 2248, 10.1002/mds.22322
Wenning, 1997, Multiple system atrophy: a review of 203 pathologically proven cases, Mov. Disord., 12, 133, 10.1002/mds.870120203
Stamelou, 2016, A review of treatment options for progressive supranuclear palsy, CNS Drugs, 30, 629, 10.1007/s40263-016-0347-2
Lamb, 2016, Progressive supranuclear palsy and corticobasal degeneration: pathophysiology and treatment options, Curr. Treat. Options Neurol., 18, 42, 10.1007/s11940-016-0422-5
Fanciulli, 2015, Multiple-system atrophy, N. Engl. J. Med., 372, 249, 10.1056/NEJMra1311488
Poewe, 2015, Therapeutic advances in multiple system atrophy and progressive supranuclear palsy, Mov. Disord., 30, 1528, 10.1002/mds.26334
Marsili, 2016, Therapeutic interventions in parkinsonism: corticobasal degeneration, Park. Relat. Disord., 22, S96, 10.1016/j.parkreldis.2015.09.023
Rohrer, 2018, Symptomatic therapy of multiple system atrophy, Auton. Neurosci., 211, 26, 10.1016/j.autneu.2017.10.006
Meissner, 2016, Outcome of deep brain stimulation in slowly progressive multiple system atrophy: a clinico-pathological series and review of the literature, Park. Relat. Disord., 24, 69, 10.1016/j.parkreldis.2016.01.005
Wen, 2017, A phase 2 study of Nelotanserin, a novel 5HT2A Receptor inverse agonist
Murata, 2018, Adjunct zonisamide to levodopa for DLB parkinsonism: a randomized double-blind phase 2 study, Neurology, 90, e664, 10.1212/WNL.0000000000005010