Abstract:
This short editorial provides our point of view of the first EURO TAU meeting focusing on tau diagnostics and clinical studies. We cover postmortem analyses toward the identification of new biomarkers, tau imaging as a diagnostic biomarker, cerebrospinal fluid (CSF) sampling with emphasis on tau fragments, blood tests and genetic evaluations for sporadic cases, treatment aspects, drug development and points for future developments toward disease modification of devastating tauopathies.
Introduction
Recently, Tong Guo, Wendy Noble, and Diane P. Hanger published a comprehensive review about the roles of tau protein in health and disease (Guo et al. 2017). In combination with previous reviews, including the Alzheimer's Association Research Roundtable Fall 2015—Tau: From research to clinical development (Holtzman et al. 2016) and the excellent first EURO TAU meeting, Lille, France, April 2017 (http://lucbuee.fr/crbst_10.html), we are now ideally positioned to discuss advances and future projections for tau diagnostics and clinical trials. This short review presents our own perspectives, trying to portray an objective view (Gozes 2002; Matsuoka et al. 2007; Vulih-Shultzman et al. 2007; Matsuoka et al. 2008; Shiryaev et al. 2009; Gozes 2010a, 2010b; Knake et al. 2010; Shiryaev et al. 2010; Stamelou et al. 2010; Gozes 2011; Hoglinger et al. 2011; Idan-Feldman et al. 2012; Jouroukhin et al. 2012; Jouroukhin et al. 2013; Boxer et al. 2014; Gozes et al. 2014; Hoglinger et al. 2014; Magen et al. 2014; Schirer et al. 2014; Tolosa et al. 2014; Kouri et al. 2015, Sun et al. 2015, Levin et al. 2016, Stamelou and Hoglinger 2016; Boxer et al. 2017; Gozes 2017; Hansson et al. 2017; Hoglinger et al. 2017; Hoglund et al. 2017; Respondek et al. 2017).
Key Terms of Reference/Definitions
The article discusses key terms regarding postmortem analyses, tau imaging as a diagnostic biomarker ([18F]AV1451 and of [18F]THK5351); cerebrospinal fluid (CSF) sampling with emphasis on tau fragments (N-terminal fragments, NTF and C-terminal fragments, CTF); blood tests; genetic evaluations for sporadic cases; treatment options and drug development (NAP=davunetide=CP201, O-GlcNAcase inhibitors, tau vaccine AADvac1) toward future clinical trials.
Round Table Discussion and Questions Related to Tau Diagnostics and Clinical Studies
Postmortem Analyses, Toward Biomarkers
From the meeting perspective, recent advances at the diagnostic postmortem level were discussed. Irina Alafuzoff discussed incidence and distribution of tau pathology as well as amyloid plaques, TDP43 and α-synuclein pathologies, in various ages and clinical conditions. One of her major findings suggests that altered proteins are common in the brains of cognitively unimpaired aged subjects. This finding should be further considered while developing diagnostic biomarkers, particularly for identifying subjects at early stages of neurodegenerative diseases (Elobeid et al. 2016). Johannes Attems continued with the role of tau in the multi-morbid old brain, showing co-existence of pathologies and potential additive effects in the human brain - cross-seeding and molecular interactions between disease-associated aggregates/proteins, exemplifying interactions of pathological proteins in neurodegenerative diseases (Attems 2017). These postmortem analyses can be carried further in an automated mode for quantifying pathology in multiple brain regions to allow the identification of novel clinico-pathological phenotypes for the improvement of stratification of clinical cohorts according to underlying pathologies (Walker et al. 2017). Alberto Rabano discussed tau immunoreactive nuclear indentations in the entorhinal cortex of early Alzheimer's disease (AD) pathology. Specifically, Rabano's recent results suggest that tau immunoreactive nuclear indentations can become a possible indicator of increased total tau and/or increased 4R/3R-tau ratio in the affected neurons (Fernandez-Nogales et al. 2017) . One of the coauthors of this article (Gunter Hoglinger) discussed the distinct postmortem pathology of the primary tauopathies corticobasal degeneration (CBD) and progressive supranuclear palsy (PSP) (Hoglinger et al. 2017) showing astrocytic plaques and tafts, respectively (Stamelou et al. 2010; Levin et al. 2016).
Tau Imaging as a Diagnostic Biomarker
This topic was not extensively discussed but should be mentioned in the context of diagnosis. Indeed, recent studies are now showing hierarchical organization of tau and amyloid deposits in the cerebral cortex of the elderly brain. These deposits generally display well-defined hierarchical cortical relationships as well as overlaps between the principal clusters of both pathologic alterations in the heteromodal association regions, representing systematic, large-scale mechanisms of early AD pathology (Sepulcre et al. 2017). [18F]AV1451, used to label tau tangles in frontotemporal dementia, with the V337M MAPT mutation, showed a significant correlation between the degree of regional MRI brain atrophy and the extent of binding in the proband (Spina et al. 2017). It should be noted that [18F]AV1451 interacts also with neuromelanin in the midbrain, and may therefore be a measure of the pigmented dopaminergic neuronal count in the substantia nigra (Hansen et al. 2016). Furthermore, a difference was noted for [18F]AV1451 imaging results in AD and PSP and a cautionary note was added regarding potential off-target labelling (Passamonti et al. 2017). Imaging with [18F]THK5317 (tau deposition) and [18F]FDG (glucose metabolism) highlighted the heterogeneous propagation of tau pathology among patients with symptomatic AD, in contrast to the homogeneous changes seen in glucose metabolism, which better tracked clinical progression (Chiotis et al. 2017). However, high levels of [18F]THK5351 retention in brain regions thought to contain negligible concentrations of tau paired helical filaments raise questions about the interpretation of the positron emission tomography (PET) signals, particularly given interactions between quinolone derivatives, as with [18F]THK5317 and monoamine oxidase B (MAO-B). Thus, the interpretation of [18F]THK5351 PET images, with respect to tau, is confounded by the high MAO-B availability across the entire brain (Ng et al. 2017), paving the path to future studies.
Recommendation
improved tau imaging markers are required to standardize the methodology and apply it to clinical use, with new generation tracers such as Merck’s MK-6240 (Walji et al. 2016) or Piramal’s PI-2620 (presented at the AD/PD meeting, 2017, http://www.acimmune.com/content/images/ADPD%202017%20_PI2620_Seibyl_final.pdf).
Cerebrospinal Fluid (CSF) Sampling with Emphasis on Tau Fragments, Blood Tests and Genetic Evaluations for Sporadic Cases:
Tau measured in CSF is an established biomarker for AD; increased CSF concentrations of total tau (T-tau) are thought to reflect neurodegeneration whereas highly phosphorylated tau (P-tau) is believed to reflect tangle pathology. Importantly, despite the observation that primary tauopathies present severe neurodegeneration and tau pathology, CSF T-tau and P-tau show no clear change (Masters et al. 2015).This may be due to the fact that current tau immunoassays are based on antibodies binding to the mid region of tau and may miss pathogenic tau due to truncation or masking during oligomer/aggregate formation. Recent studies suggest that CSF tau consists of several protein fragments (Meredith et al. 2013, Barthelemy et al. 2016), and metabolic processing of tau into N-, mid and C-terminal fragments may also be key for secretion, aggregation and spreading pathology of tau, and may differ between the tauopathies (see below). Furthermore, the potential of tau oligomers or cis-tau variants (Kondo et al. 2015) as specific biomarkers for tauopathies has not been explored. Levels of tau oligomers have been shown to rise in both the CSF (Sengupta et al. 2017) and plasma (Kolarova et al. 2017) of patients with AD compared to healthy controls.
Recent data suggest that tau is actively secreted into the extracellular space and total tau along with tau fragments have been identified in the interstitial fluid (ISF) (Yanamandra et al. 2017). This suggests that some tau fragments may be actively secreted presenting a potential to give additive biomarker information. In this respect, proteolysis of tau is mediated by a range of different proteases, some of which break down the tertiary paper clip like structure of tau (Wang and Mandelkow 2015) resulting in exacerbated tau aggregation (Sokolow et al. 2015). The resulting proteolytic fragments have different characteristics dependent on the site of cleavage within tau and also depending on the inclusion of the microtubule-binding domain (Yin and Kuret 2006), (Wray et al. 2008, Matsumoto et al. 2015). One CTF, termed tau35 (Arai et al. 2004; Wray et al. 2008), that is specific to CBD and PSP, induced cognitive deficits when expressed at low levels in the Tau35 mouse, which could be reversed with sodium-4-phenylbutyrate treatment (Bondulich et al. 2016). Notably, sodium 4-phenylbutyrate is a well-known histone deacetylase inhibitor and chromatin modification through histone acetylation is a molecular pathway involved in the regulation of transcription underlying memory storage. Another transgenic mouse expressing tau151-391 developed hyperphosphorylated and aggregated tau species, deficits in locomotor activity, and had a reduced lifespan (Zimova et al. 2016). Recently, a study characterizing tau CTFs in 12 cortical samples from both control and late Braak stage AD, led to the identification of 21 novel tau fragments (Derisbourg et al. 2015).
Proteolytic fragments of tau have been found in the CSF and plasma (Henriksen et al. 2013; Henriksen et al. 2015; Inekci et al. 2015) of patients with different tauopathies, presenting these fragments as potential novel biomarkers for disease progression. Recent data have shown that fragments of tau are able to cross the blood-brain barrier with different rates, depending on the precise amino acid sequence (Banks et al. 2016). The CSF tau profile is composed entirely of tau fragments rather than full-length protein, with different fragmentation patterns dependent on disease state and progression (Johnson et al. 1997; Meredith et al. 2013). CSF tau CTF and NTF levels are decreased in PSP, while increases were observed in AD (Wagshal et al. 2015). There is much interest in the use of CSF fragments of tau as a biomarker, with data being presented by Kina Höglund showing that the asparagine endopeptidase mediated-NTF, tau1-368 (Zhang et al. 2014) could potentially act as an AD specific biomarker.
Tau protein can also be measured in blood, but the potential for identification of AD or neurodegeneration is lower for blood tau than for CSF tau (Mattsson et al. 2016). This lack of correlation between blood and CSF might be due to the “wrong” pool or the fragment of tau that is being analyzed.
An intriguing finding is that exosomes, which are secreted in body fluids such as CSF and serum, contain tau protein (Winston et al. 2016). Exosomes are believed to be a way for cells to communicate, and thus, may present a potential mechanism for cell-to-cell propagation of tau aggregates. Some studies indicate that tau from isolated brain-specific exosomes in blood may have value as a biomarker for brain disorders such as AD (Winston et al. 2016).
From the point of view of the authors of this article, Kina Höglund expands the cerebrospinal fluid endopeptidome (Hansson et al. 2017; Hoglund et al. 2017), while Illana Gozes studies potential blood biomarkers using genome-wide transcriptomic profiling and bioinformatics data mining (Hadar et al. 2016). A proteomic approach identified the tau-interacting activity-dependent neuroprotective protein (ADNP) (Oz et al. 2012; Schirer et al. 2014; Ivashko-Pachima et al. 2017), as the only protein down-regulated in serum samples from early AD patients (Yang et al. 2012). These studies were corroborated showing that blood borne expression of ADNP is correlated with premorbid intelligence, AD pathology, and clinical stage. Age adjustment showed significant associations between: 1) higher premorbid intelligence and greater serum ADNP, and 2) greater cortical amyloid and lower ADNP mRNA. Furthermore, increased ADNP mRNA levels were observed in patients ranging from mild cognitive impairment (MCI) to AD dementia, suggesting additional, tau-related blood markers (Malishkevich et al. 2016).
Thibaud Lebouvier discussed clinical aspects aiming to identify genetic markers and gene signatures associated with CSF biomarker levels of t-tau, p-tau181, and with the two ratios t-tau/Aβ1-42 and p-tau181/Aβ1-42 in the context of progression from MCI to AD, and to identify a panel of genetic markers that can predict CSF biomarker p-tau181/Aβ1-42 ratio with consideration of APOE4 stratification. The group analyzed genome-wide the AD Neuroimaging Initiative (ADNI) dataset. They identified a panel of five SNPs, rs6766238, rs1143960, rs1249963, rs11975968, and rs4836493, that are predictive for p-tau181/Aβ1-42 ratio (high/low) with a sensitivity of 66% and a specificity of 70% (AUC 0.74). These results suggest that a panel of SNPs is a potential prognostic biomarker in ApoE4-negative MCI patients (Sun et al. 2015). Furthermore, from a genetic point of view, Gunter Hoglinger showed that the tau encoding gene MAPT, is the major risk gene for PSP (Hoglinger et al. 2011) and CBD (Kouri et al. 2015).
Recommendation
large cohorts and longitudinal studies are needed to verify potential tau fragments or other proteins/peptides as specific biomarkers in CSF and perhaps in other body fluids, e.g. blood.
Treatment Options
Treatment options for the primary tauopathy, PSP, were discussed by Gunter Hoglinger, including levodopa, amantadine, amitriptyline, zolpidem, CoQ10 and Botilinum toxin A, all extensively reviewed in the published literature (Stamelou and Hoglinger 2016).
Drug Development
Table 1 shows ongoing clinical trials directed at tau. Meanwhile, there were a few failures and some hope.
The most recent failure was with Leuco-methylthioninium bis (hydromethanesulfonate; LMTM), a stable reduced form of the methylthioninium moiety, acting as a selective inhibitor of tau protein aggregation both in vitro and in transgenic mouse models, which was tested in patients with mild to moderate AD (Gauthier et al. 2016). It is possible that drug administration was too late in the disease process, and unable to reverse previously accumulated cell damage. An additional previous failure was with the GSK-3 inhibitor tideglusib (Dominguez et al. 2012), for example, in AD (del del Ser et al. 2013) and PSP (Tolosa et al. 2014). Interestingly, while there were no physical or behavioral benefits for tideglusib, it did reduce progression of brain atrophy (Hoglinger et al. 2014).
In contrast to PSP (Boxer et al. 2014), intranasal NAP (davunetide, AL-108) showed efficacy in other indications, namely MCI, significantly increasing cognitive scores (Gozes et al. 2009; Morimoto et al. 2013) and in cognitive impairment associated with schizophrenia, significantly protecting functional activity and brain matter (Javitt et al. 2012; Jarskog et al. 2013). NAP (now called CP201) is poised for further clinical trials in the ADNP syndrome (Gozes et al. 2015, 2017a, b), within the autism spectrum disorders (http://www.coronisns.com/Pipeline/575/CP201-for-ADNP-syndrome). In this respect, ADNP through its NAP (octapeptide snippet, NAPVSIPQ) binds to microtubule end binding proteins, which in turn bind to tau and dramatically enhance tau interaction with the microtubules (Ivashko-Pachima et al. 2017), protecting against tauopathy associated with ADNP deficiency (Vulih-Shultzman et al. 2007) and enhancing synaptic plasticity (Oz et al. 2014). These results were presented by Illana Gozes at the Eurotau meeting.
Lastly, Dirk Beher presented a future clinical trial in PSP with O-GlcNAcase inhibitors with supporting evidence from preclinical trials [e.g. (Yuzwa et al. 2014; Hastings et al. 2017)] and Michal Novak discussed the safety and immunogenicity of the tau vaccine AADvac1, toward future clinical trials (Novak et al. 2017).
Recommendation
results from clinical trials currently underway will help to inform and focus the development of new tau-targeting therapies.
Discussion and Future Directions
The discussion focused on three major issues, improving early diagnosis, choosing the right cohort with relevant clinical endpoints and designing/developing drugs.
In terms of early diagnostics to provide for better treatment modalities, Johannes Attems discussed the possibility of addressing the olfactory pathology toward better, simpler early diagnosis (Attems et al. 2015).
Kina Höglund raised the issue that basic biochemical studies in brain tissue focusing on endogenous fragments/peptides of tau is one way forward to identify novel tau biomarkers. For clinical validation, it is important to have access to already established biomarkers such as CSF Aβ42, total tau, p-tau (181) as well as tau and Aβ PET imaging and MRI.
It was further suggested to take advantage of the established biomarkers to exclude, for example, patients with amyloid pathology, either based on amyloid PET or CSF Aβ42. Further studies to examine the profile of neurofilament light chain in blood in various tauopathies may also enable a screening tool where exclusion or inclusion criteria can be applied.
Illana Gozes raised the possibility of developing more/better blood biomarkers (Hadar et al. 2016; Malishkevich et al. 2016) and emphasized sex differences, both in diagnostics as well as in clinical trial design and statistical analysis of the outcomes (Malishkevich et al. 2015).
With Dirk Beher (who led the discussion) starting a clinical trial in PSP [as well as the trials outlined in Table 1(Boxer et al. 2017)], the issue of possible success was raised, and early mild cognitive impairment as a promising indication was suggested, targeting disease before extensive pathology has accumulated (Morimoto et al. 2013) (Illana Gozes).
In terms of drug design and development, the idea of multi-target drugs or drug mixtures was discussed for future pivotal meeting/trials.
References
Arai T, Ikeda K, Akiyama H, Nonaka T, Hasegawa M, Ishiguro K, Iritani S, Tsuchiya K, Iseki E, Yagishita S, Oda T, Mochizuki A (2004) Identification of amino-terminally cleaved tau fragments that distinguish progressive supranuclear palsy from corticobasal degeneration. Ann Neurol 55:72–79
Attems J (2017) The multi-morbid old brain. Acta neuropathologica.
Attems J, Walker L, Jellinger KA (2015) Olfaction and Aging: A Mini-Review. Gerontology 61:485–490
Banks WA, Kovac A, Majerova P, Bullock KM, Shi M, Zhang J (2016) Tau Proteins Cross the Blood-Brain Barrier. J Alzheim Dis 55:411–419
Barthelemy NR, Gabelle A, Hirtz C, Fenaille F, Sergeant N, Schraen-Maschke S, Vialaret J, Buee L, Junot C, Becher F, Lehmann S (2016) Differential Mass Spectrometry Profiles of Tau Protein in the Cerebrospinal Fluid of Patients with Alzheimer's Disease, Progressive Supranuclear Palsy, and Dementia with Lewy Bodies. J Alzheim Dis 51:1033–1043
Bondulich MK, Guo T, Meehan C, Manion J, Rodriguez Martin T, Mitchell JC, Hortobagyi T, Yankova N, Stygelbout V, Brion JP, Noble W, Hanger DP (2016) Tauopathy induced by low level expression of a human brain-derived tau fragment in mice is rescued by phenylbutyrate. Brain: J Neurol.
Boxer AL, Lang AE, Grossman M, Knopman DS, Miller BL, Schneider LS, Doody RS, Lees A, Golbe LI, Williams DR, Corvol JC, Ludolph A, Burn D, Lorenzl S, Litvan I, Roberson ED, Hoglinger GU, Koestler M, Jack CR, Jr., Van Deerlin V, Randolph C, Lobach IV, Heuer HW, Gozes I, Parker L, Whitaker S, Hirman J, Stewart AJ, Gold M, Morimoto BH, Investigators AL (2014) Davunetide in patients with progressive supranuclear palsy: a randomised, double-blind, placebo-controlled phase 2/3 trial. Lancet Neurol 13:676-685.
Boxer AL, Yu JT, Golbe LI, Litvan I, Lang AE, Hoglinger GU (2017) Advances in progressive supranuclear palsy: new diagnostic criteria, biomarkers, and therapeutic approaches. Lancet Neurol 16:552–563
Chiotis K, Saint-Aubert L, Rodriguez-Vieitez E, Leuzy A, Almkvist O, Savitcheva I, Jonasson M, Lubberink M, Wall A, Antoni G, Nordberg A (2017) Longitudinal changes of tau PET imaging in relation to hypometabolism in prodromal and Alzheimer's disease dementia. Mol Psychiatry.
del Ser T, Steinwachs KC, Gertz HJ, Andres MV, Gomez-Carrillo B, Medina M, Vericat JA, Redondo P, Fleet D, Leon T (2013) Treatment of Alzheimer's disease with the GSK-3 inhibitor tideglusib: a pilot study. J Alzheim Dis 33:205–215
Derisbourg M, Leghay C, Chiappetta G, Fernandez-Gomez FJ, Laurent C, Demeyer D, Carrier S, Buee-Scherrer V, Blum D, Vinh J, Sergeant N, Verdier Y, Buee L, Hamdane M (2015) Role of the Tau N-terminal region in microtubule stabilization revealed by new endogenous truncated forms. Sci Rep 5:9659
Dominguez JM, Fuertes A, Orozco L, del Monte-Millan M, Delgado E, Medina M (2012) Evidence for irreversible inhibition of glycogen synthase kinase-3beta by tideglusib. J Biol Chem 287:893–904
Elobeid A, Libard S, Leino M, Popova SN, Alafuzoff I (2016) Altered Proteins in the Aging Brain. J Neuropathol Exp Neurol 75:316–325
Fernandez-Nogales M, Santos-Galindo M, Merchan-Rubira J, Hoozemans JJM, Rabano A, Ferrer I, Avila J, Hernandez F, Lucas JJ (2017) Tau-positive nuclear indentations in P301S tauopathy mice. Brain Pathol 27:314–322
Gauthier S, Feldman HH, Schneider LS, Wilcock GK, Frisoni GB, Hardlund JH, Moebius HJ, Bentham P, Kook KA, Wischik DJ, Schelter BO, Davis CS, Staff RT, Bracoud L, Shamsi K, Storey JM, Harrington CR, Wischik CM (2016) Efficacy and safety of tau-aggregation inhibitor therapy in patients with mild or moderate Alzheimer's disease: a randomised, controlled, double-blind, parallel-arm, phase 3 trial. Lancet 388:2873–2884
Gozes I (2002) Tau as a drug target in Alzheimer's disease. J Mol Neurosci 19:337–338
Gozes I (2010a) Tau pathology and future therapeutics. Curr Alzheimer Res 7:685–696
Gozes I (2010b) Tau pathology: predictive diagnostics, targeted preventive and personalized medicine and application of advanced research in medical practice. EPMA J 1:305–316
Gozes I (2011) Microtubules (tau) as an emerging therapeutic target: NAP (davunetide). Curr Pharm Des 17:3413–3417
Gozes I (2017) Sexual divergence in activity-dependent neuroprotective protein impacting autism, schizophrenia, and Alzheimer's disease. J Neurosci Res 95:652–660
Gozes I, Stewart A, Morimoto B, Fox A, Sutherland K, Schmeche D (2009) Addressing Alzheimer's disease tangles: from NAP to AL-108. Curr Alzheimer Res 6:455–460
Gozes I, Iram T, Maryanovsky E, Arviv C, Rozenberg L, Schirer Y, Giladi E, Furman-Assaf S (2014) Novel tubulin and tau neuroprotective fragments sharing structural similarities with the drug candidate NAP (Davuentide). J Alzheim Dis 40(Suppl 1):S23–S36
Gozes I, Helsmoortel C, Vandeweyer G, Van der Aa N, Kooy F, Sermone SB (2015) The Compassionate Side of Neuroscience: Tony Sermone's Undiagnosed Genetic Journey--ADNP Mutation. J Mol Neurosci 56:751-757.
Gozes I, Patterson MC, Van Dijck A, Kooy RF, Peeden JN, Eichenberger JA, Zawacki-Downing A, Bedrosian-Sermone S (2017a) The Eight and a Half Year Journey of Undiagnosed AD: Gene Sequencing and Funding of Advanced Genetic Testing Has Led to Hope and New Beginnings. Front Endocrinol 8:107.
Gozes I, Van Dijck A, Hacohen-Kleiman G, Grigg I, Karmon G, Giladi E, Eger M, Gabet Y, Pasmanik-Chor M, Cappuyns E, Elpeleg O, Kooy RF, Bedrosian-Sermone S (2017b) Premature primary tooth eruption in cognitive/motor-delayed ADNP-mutated children. Transl Psychiatry 7:e1043.
Guo T, Noble W, Hanger DP (2017) Roles of tau protein in health and disease. Acta Neuropathol 133:665–704
Hadar A, Milanesi E, Squassina A, Niola P, Chillotti C, Pasmanik-Chor M, Yaron O, Martasek P, Rehavi M, Weissglas-Volkov D, Shomron N, Gozes I, Gurwitz D (2016) RGS2 expression predicts amyloid-beta sensitivity, MCI and Alzheimer's disease: genome-wide transcriptomic profiling and bioinformatics data mining. Transl Psychiatry 6:e909
Hansen AK, Knudsen K, Lillethorup TP, Landau AM, Parbo P, Fedorova T, Audrain H, Bender D, Ostergaard K, Brooks DJ, Borghammer P (2016) In vivo imaging of neuromelanin in Parkinson's disease using 18F-AV-1451 PET. Brain J Neurol 139:2039–2049
Hansson KT, Skillback T, Pernevik E, Kern S, Portelius E, Hoglund K, Brinkmalm G, Holmen-Larsson J, Blennow K, Zetterberg H, Gobom J (2017) Expanding the cerebrospinal fluid endopeptidome. Proteomics 17
Hastings NB, Wang X, Song L, Butts BD, Grotz D, Hargreaves R, Fred Hess J, Hong KK, Huang CR, Hyde L, Laverty M, Lee J, Levitan D, Lu SX, Maguire M, Mahadomrongkul V, McEachern EJ, Ouyang X, Rosahl TW, Selnick H, Stanton M, Terracina G, Vocadlo DJ, Wang G, Duffy JL, Parker EM, Zhang L (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
Henriksen K, Wang Y, Sorensen MG, Barascuk N, Suhy J, Pedersen JT, Duffin KL, Dean RA, Pajak M, Christiansen C, Zheng Q, Karsdal MA (2013) An enzyme-generated fragment of tau measured in serum shows an inverse correlation to cognitive function. PLoS One 8:e64990.
Henriksen K, Byrjalsen I, Christiansen C, Karsdal MA (2015) Relationship between serum levels of tau fragments and clinical progression of Alzheimer's disease. J Alzheim Dis 43:1331–1341
Hoglinger GU, Melhem NM, Dickson DW, Sleiman PM, Wang LS, Klei L, Rademakers R, de Silva R, Litvan I, Riley DE, van Swieten JC, Heutink P, Wszolek ZK, Uitti RJ, Vandrovcova J, Hurtig HI, Gross RG, Maetzler W, Goldwurm S, Tolosa E, Borroni B, Pastor P, Group PSPGS, Cantwell LB, Han MR, Dillman A, van der Brug MP, Gibbs JR, Cookson MR, Hernandez DG, Singleton AB, Farrer MJ, Yu CE, Golbe LI, Revesz T, Hardy J, Lees AJ, Devlin B, Hakonarson H, Muller U, Schellenberg GD (2011) Identification of common variants influencing risk of the tauopathy progressive supranuclear palsy. Nat Genet 43:699-705.
Hoglinger GU, Huppertz HJ, Wagenpfeil S, Andres MV, Belloch V, Leon T, Del Ser T, Investigators TM (2014) Tideglusib reduces progression of brain atrophy in progressive supranuclear palsy in a randomized trial. Mov Disord: Off J Mov Disord Soc 29:479–487
Hoglinger GU, Respondek G, Stamelou M, Kurz C, Josephs KA, Lang AE, Mollenhauer B, Muller U, Nilsson C, Whitwell JL, Arzberger T, Englund E, Gelpi E, Giese A, Irwin DJ, Meissner WG, Pantelyat A, Rajput A, van Swieten JC, Troakes C, Antonini A, Bhatia KP, Bordelon Y, Compta Y, Corvol JC, Colosimo C, Dickson DW, Dodel R, Ferguson L, Grossman M, Kassubek J, Krismer F, Levin J, Lorenzl S, Morris HR, Nestor P, Oertel WH, Poewe W, Rabinovici G, Rowe JB, Schellenberg GD, Seppi K, van Eimeren T, Wenning GK, Boxer AL, Golbe LI, Litvan I, Movement Disorder Society-endorsed PSPSG (2017) Clinical diagnosis of progressive supranuclear palsy: The movement disorder society criteria. Mov Disord: Off J Mov Disord Soc
Hoglund K, Kern S, Zettergren A, Borjesson-Hansson A, Zetterberg H, Skoog I, Blennow K (2017) Preclinical amyloid pathology biomarker positivity: effects on tau pathology and neurodegeneration. Transl Psychiatry 7:e995
Holtzman DM, Carrillo MC, Hendrix JA, Bain LJ, Catafau AM, Gault LM, Goedert M, Mandelkow E, Mandelkow EM, Miller DS, Ostrowitzki S, Polydoro M, Smith S, Wittmann M, Hutton M (2016) Tau: From research to clinical development. Alzheimer's & dementia : the journal of the Alzheimer's Association 12:1033–1039
Idan-Feldman A, Ostritsky R, Gozes I (2012) Tau and caspase 3 as targets for neuroprotection. Int J Alzheimers Dis 2012:493670
Inekci D, Henriksen K, Linemann T, Karsdal MA, Habib A, Bisgaard C, Eriksen FB, Vilholm OJ (2015) Serum fragments of Tau for the differential diagnosis of Alzheimer's disease. Curr Alzheimer Res
Ivashko-Pachima Y, Sayas CL, Malishkevich A, Gozes I (2017) ADNP/NAP dramatically increase microtubule end-binding protein-Tau interaction: a novel avenue for protection against tauopathy. Mol Psychiatry 22:1335–1344
Jarskog LF, Dong Z, Kangarlu A, Colibazzi T, Girgis RR, Kegeles LS, Barch DM, Buchanan RW, Csernansky JG, Goff DC, Harms MP, Javitt DC, Keefe RS, McEvoy JP, McMahon RP, Marder SR, Peterson BS, Lieberman JA (2013) Effects of davunetide on N-acetylaspartate and choline in dorsolateral prefrontal cortex in patients with schizophrenia. Neuropsychopharmacol : Offi Publ Am Collegef Neuropsychopharmacol 38:1245–1252
Javitt DC, Buchanan RW, Keefe RS, Kern R, McMahon RP, Green MF, Lieberman J, Goff DC, Csernansky JG, McEvoy JP, Jarskog F, Seidman LJ, Gold JM, Kimhy D, Nolan KS, Barch DS, Ball MP, Robinson J, Marder SR (2012) Effect of the neuroprotective peptide davunetide (AL-108) on cognition and functional capacity in schizophrenia. Schizophr Res 136:25–31
Johnson GV, Seubert P, Cox TM, Motter R, Brown JP, Galasko D (1997) The tau protein in human cerebrospinal fluid in Alzheimer's disease consists of proteolytically derived fragments. J Neurochem 68:430–433
Jouroukhin Y, Ostritsky R, Gozes I (2012) D-NAP prophylactic treatment in the SOD mutant mouse model of amyotrophic lateral sclerosis: review of discovery and treatment of tauopathy. J Mol Neurosci 48:597–602
Jouroukhin Y, Ostritsky R, Assaf Y, Pelled G, Giladi E, Gozes I (2013) NAP (davunetide) modifies disease progression in a mouse model of severe neurodegeneration: protection against impairments in axonal transport. Neurobiol Dis 56:79–94
Knake S, Belke M, Menzler K, Pilatus U, Eggert KM, Oertel WH, Stamelou M, Hoglinger GU (2010) In vivo demonstration of microstructural brain pathology in progressive supranuclear palsy: a DTI study using TBSS. Mov Disord: Off J Mov Disord Soc 25:1232–1238
Kolarova M, Sengupta U, Bartos A, Ricny J, Kayed R (2017) Tau Oligomers in Sera of Patients with Alzheimer's Disease and Aged Controls. J Alzheim Dis 58:471–478
Kondo A, Shahpasand K, Mannix R, Qiu J, Moncaster J, Chen CH, Yao Y, Lin YM, Driver JA, Sun Y, Wei S, Luo ML, Albayram O, Huang P, Rotenberg A, Ryo A, Goldstein LE, Pascual-Leone A, McKee AC, Meehan W, Zhou XZ, Lu KP (2015) Antibody against early driver of neurodegeneration cis P-tau blocks brain injury and tauopathy. Nature 523:431–436
Kouri N, Ross OA, Dombroski B, Younkin CS, Serie DJ, Soto-Ortolaza A, Baker M, Finch NC, Yoon H, Kim J, Fujioka S, McLean CA, Ghetti B, Spina S, Cantwell LB, Farlow MR, Grafman J, Huey ED, Ryung Han M, Beecher S, Geller ET, Kretzschmar HA, Roeber S, Gearing M, Juncos JL, Vonsattel JP, Van Deerlin VM, Grossman M, Hurtig HI, Gross RG, Arnold SE, Trojanowski JQ, Lee VM, Wenning GK, White CL, Hoglinger GU, Muller U, Devlin B, Golbe LI, Crook J, Parisi JE, Boeve BF, Josephs KA, Wszolek ZK, Uitti RJ, Graff-Radford NR, Litvan I, Younkin SG, Wang LS, Ertekin-Taner N, Rademakers R, Hakonarsen H, Schellenberg GD, Dickson DW (2015) Genome-wide association study of corticobasal degeneration identifies risk variants shared with progressive supranuclear palsy. Nat Commun 6:7247.
Levin J, Kurz A, Arzberger T, Giese A, Hoglinger GU (2016) The Differential Diagnosis and Treatment of Atypical Parkinsonism. Dtsch Arztebl int 113:61–69
Magen I, Ostritsky R, Richter F, Zhu C, Fleming SM, Lemesre V, Stewart AJ, Morimoto BH, Gozes I, Chesselet MF (2014) Intranasal NAP (davunetide) decreases tau hyperphosphorylation and moderately improves behavioral deficits in mice overexpressing alpha-synuclein. Pharmacol Res Perspect 2:e00065
Malishkevich A, Amram N, Hacohen-Kleiman G, Magen I, Giladi E, Gozes I (2015) Activity-dependent neuroprotective protein (ADNP) exhibits striking sexual dichotomy impacting on autistic and Alzheimer's pathologies. Transl Psychiatry 5:e501
Malishkevich A, Marshall GA, Schultz AP, Sperling RA, Aharon-Peretz J, Gozes I (2016) Blood-Borne Activity-Dependent Neuroprotective Protein (ADNP) is Correlated with Premorbid Intelligence, Clinical Stage, and Alzheimer's Disease Biomarkers. J Alzheim Dis 50:249–260
Masters CL, Bateman R, Blennow K, Rowe CC, Sperling RA, Cummings JL (2015) Alzheimer's disease. Nat Rev Dis Primers 1:15056
Matsumoto SE, Motoi Y, Ishiguro K, Tabira T, Kametani F, Hasegawa M, Hattori N (2015) The twenty-four KDa C-terminal tau fragment increases with aging in tauopathy mice: implications of prion-like properties. Hum Mol Genet 24:6403–6416
Matsuoka Y, Gray AJ, Hirata-Fukae C, Minami SS, Waterhouse EG, Mattson MP, LaFerla FM, Gozes I, Aisen PS (2007) Intranasal NAP administration reduces accumulation of amyloid peptide and tau hyperphosphorylation in a transgenic mouse model of Alzheimer's disease at early pathological stage. J Mol Neurosci 31:165–170
Matsuoka Y, Jouroukhin Y, Gray AJ, Ma L, Hirata-Fukae C, Li HF, Feng L, Lecanu L, Walker BR, Planel E, Arancio O, Gozes I, Aisen PS (2008) A neuronal microtubule-interacting agent, NAPVSIPQ, reduces tau pathology and enhances cognitive function in a mouse model of Alzheimer's disease. J Pharmacol Exp Ther 325:146–153
Mattsson N, Zetterberg H, Janelidze S, Insel PS, Andreasson U, Stomrud E, Palmqvist S, Baker D, Tan Hehir CA, Jeromin A, Hanlon D, Song L, Shaw LM, Trojanowski JQ, Weiner MW, Hansson O, Blennow K, Investigators A (2016) Plasma tau in Alzheimer disease. Neurology 87:1827–1835
Meredith JE Jr, Sankaranarayanan S, Guss V, Lanzetti AJ, Berisha F, Neely RJ, Slemmon JR, Portelius E, Zetterberg H, Blennow K, Soares H, Ahlijanian M, Albright CF (2013) Characterization of novel CSF Tau and ptau biomarkers for Alzheimer's disease. PLoS One 8:e76523
Morimoto BH, Schmechel D, Hirman J, Blackwell A, Keith J, Gold M, Study AL (2013) A double-blind, placebo-controlled, ascending-dose, randomized study to evaluate the safety, tolerability and effects on cognition of AL-108 after 12 weeks of intranasal administration in subjects with mild cognitive impairment. Dement Geriatr Cogn Disord 35:325–336
Ng KP, Pascoal TA, Mathotaarachchi S, Therriault J, Kang MS, Shin M, Guiot MC, Guo Q, Harada R, Comley RA, Massarweh G, Soucy JP, Okamura N, Gauthier S, Rosa-Neto P (2017) Monoamine oxidase B inhibitor, selegiline, reduces 18F-THK5351 uptake in the human brain. Alzheimers Res Ther 9:25
Novak P, Schmidt R, Kontsekova E, Zilka N, Kovacech B, Skrabana R, Vince-Kazmerova Z, Katina S, Fialova L, Prcina M, Parrak V, Dal-Bianco P, Brunner M, Staffen W, Rainer M, Ondrus M, Ropele S, Smisek M, Sivak R, Winblad B, Novak M (2017) Safety and immunogenicity of the tau vaccine AADvac1 in patients with Alzheimer's disease: a randomised, double-blind, placebo-controlled, phase 1 trial. Lancet Neurol 16:123–134
Oz S, Ivashko-Pachima Y, Gozes I (2012) The ADNP derived peptide, NAP modulates the tubulin pool: implication for neurotrophic and neuroprotective activities. PLoS One 7:e51458
Oz S, Kapitansky O, Ivashco-Pachima Y, Malishkevich A, Giladi E, Skalka N, Rosin-Arbesfeld R, Mittelman L, Segev O, Hirsch JA, Gozes I (2014) The NAP motif of activity-dependent neuroprotective protein (ADNP) regulates dendritic spines through microtubule end binding proteins. Mol Psychiatry 19:1115–1124
Passamonti L, Vazquez Rodriguez P, Hong YT, Allinson KS, Williamson D, Borchert RJ, Sami S, Cope TE, Bevan-Jones WR, Jones PS, Arnold R, Surendranathan A, Mak E, Su L, Fryer TD, Aigbirhio FI, O'Brien JT, Rowe JB (2017) 18F-AV-1451 positron emission tomography in Alzheimer's disease and progressive supranuclear palsy. Brain J Neurol 140:781–791
Respondek G, Kurz C, Arzberger T, Compta Y, Englund E, Ferguson LW, Gelpi E, Giese A, Irwin DJ, Meissner WG, Nilsson C, Pantelyat A, Rajput A, van Swieten JC, Troakes C, Josephs KA, Lang AE, Mollenhauer B, Muller U, Whitwell JL, Antonini A, Bhatia KP, Bordelon Y, Corvol JC, Colosimo C, Dodel R, Grossman M, Kassubek J, Krismer F, Levin J, Lorenzl S, Morris H, Nestor P, Oertel WH, Rabinovici GD, Rowe JB, van Eimeren T, Wenning GK, Boxer A, Golbe LI, Litvan I, Stamelou M, Hoglinger GU, Movement Disorder Society-Endorsed PSPSG (2017) Which ante mortem clinical features predict progressive supranuclear palsy pathology? Mov Disord: Off J Mov Disord Soc
Schirer Y, Malishkevich A, Ophir Y, Lewis J, Giladi E, Gozes I (2014) Novel marker for the onset of frontotemporal dementia: early increase in activity-dependent neuroprotective protein (ADNP) in the face of Tau mutation. PLoS One 9:e87383
Sengupta U, Portelius E, Hansson O, Farmer K, Castillo-Carranza D, Woltjer R, Zetterberg H, Galasko D, Blennow K, Kayed R (2017) Tau oligomers in cerebrospinal fluid in Alzheimer's disease. Ann Clin Transl Neurol 4:226–235
Sepulcre J, Grothe MJ, Sabuncu M, Chhatwal J, Schultz AP, Hanseeuw B, El Fakhri G, Sperling R, Johnson KA (2017) Hierarchical Organization of Tau and Amyloid Deposits in the Cerebral Cortex. JAMA Neurol
Shiryaev N, Jouroukhin Y, Giladi E, Polyzoidou E, Grigoriadis NC, Rosenmann H, Gozes I (2009) NAP protects memory, increases soluble tau and reduces tau hyperphosphorylation in a tauopathy model. Neurobiol Dis 34:381–388
Shiryaev N, Jouroukhin Y, Gozes I (2010) 3R tau expression modifies behavior in transgenic mice. J Neurosci Res 88:2727–2735
Sokolow S, Henkins KM, Bilousova T, Gonzalez B, Vinters HV, Miller CA, Cornwell L, Poon WW, Gylys KH (2015) Pre-synaptic C-terminal truncated tau is released from cortical synapses in Alzheimer's disease. J Neurochem 133:368–379
Spina S, Schonhaut DR, Boeve BF, Seeley WW, Ossenkoppele R, O'Neil JP, Lazaris A, Rosen HJ, Boxer AL, Perry DC, Miller BL, Dickson DW, Parisi JE, Jagust WJ, Murray ME, Rabinovici GD (2017) Frontotemporal dementia with the V337M MAPT mutation: Tau-PET and pathology correlations. Neurology 88:758–766
Stamelou M, Hoglinger G (2016) A Review of Treatment Options for Progressive Supranuclear Palsy. CNS drugs 30:629–636
Stamelou M, de Silva R, Arias-Carrion O, Boura E, Hollerhage M, Oertel WH, Muller U, Hoglinger GU (2010) Rational therapeutic approaches to progressive supranuclear palsy. Brain J Neurol 133:1578-1590.
Sun Y, Bresell A, Rantalainen M, Hoglund K, Lebouvier T, Salter H, Alzheimer Disease Neuroimaging I (2015) An Integrated Bioinformatics Approach for Identifying Genetic Markers that Predict Cerebrospinal Fluid Biomarker p-tau181/Abeta1-42 Ratio in ApoE4-Negative Mild Cognitive Impairment Patients. J Alzheim Dis 45:1061–1076
Tolosa E, Litvan I, Hoglinger GU, Burn D, Lees A, Andres MV, Gomez-Carrillo B, Leon T, Del Ser T, Investigators T (2014) A phase 2 trial of the GSK-3 inhibitor tideglusib in progressive supranuclear palsy. Mov Disord: Off J Mov Disord Soc 29:470–478
Vulih-Shultzman I, Pinhasov A, Mandel S, Grigoriadis N, Touloumi O, Pittel Z, Gozes I (2007) Activity-dependent neuroprotective protein snippet NAP reduces tau hyperphosphorylation and enhances learning in a novel transgenic mouse model. J Pharmacol Exp Ther 323:438–449
Wagshal D, Sankaranarayanan S, Guss V, Hall T, Berisha F, Lobach I, Karydas A, Voltarelli L, Scherling C, Heuer H, Tartaglia MC, Miller Z, Coppola G, Ahlijanian M, Soares H, Kramer JH, Rabinovici GD, Rosen HJ, Miller BL, Meredith J, Boxer AL (2015) Divergent CSF tau alterations in two common tauopathies: Alzheimer's disease and progressive supranuclear palsy. J Neurol Neurosurg Psychiatry 86:244–250
Walji AM, Hostetler ED, Selnick H, Zeng Z, Miller P, Bennacef I, Salinas C, Connolly B, Gantert L, Holahan M, O'Malley S, Purcell M, Riffel K, Li J, Balsells J, OB JA, Melquist S, Soriano A, Zhang X, Ogawa A, Xu S, Joshi E, Della Rocca J, Hess FJ, Schachter J, Hesk D, Schenk D, Struyk A, Babaoglu K, Lohith TG, Wang Y, Yang K, Fu J, Evelhoch JL, Coleman PJ (2016) Discovery of 6-(Fluoro-(18)F)-3-(1H-pyrrolo[2,3-c]pyridin-1-yl)isoquinolin-5-amine ([(18)F]-MK-6240): A Positron Emission Tomography (PET) Imaging Agent for Quantification of Neurofibrillary Tangles (NFTs). J Med Chem 59:4778–4789
Walker L, McAleese KE, Johnson M, Khundakar AA, Erskine D, Thomas AJ, McKeith IG, Attems J (2017) Quantitative neuropathology: an update on automated methodologies and implications for large scale cohorts. J Neural Transm
Wang Y, Mandelkow E (2015) Tau in physiology and pathology. Nat Rev Neurosci 17:22–35
Winston CN, Goetzl EJ, Akers JC, Carter BS, Rockenstein EM, Galasko D, Masliah E, Rissman RA (2016) Prediction of conversion from mild cognitive impairment to dementia with neuronally derived blood exosome protein profile. Alzheimers Dement 3:63–72
Wray S, Saxton M, Anderton BH, Hanger DP (2008) 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 105:2343–2352
Yanamandra K, Patel TK, Jiang H, Schindler S, Ulrich JD, Boxer AL, Miller BL, Kerwin DR, Gallardo G, Stewart F, Finn MB, Cairns NJ, Verghese PB, Fogelman I, West T, Braunstein J, Robinson G, Keyser J, Roh J, Knapik SS, Hu Y, Holtzman DM (2017) Anti-tau antibody administration increases plasma tau in transgenic mice and patients with tauopathy. Sci Transl Med 9
Yang MH, Yang YH, Lu CY, Jong SB, Chen LJ, Lin YF, Wu SJ, Chu PY, Chung TW, Tyan YC (2012) Activity-dependent neuroprotector homeobox protein: A candidate protein identified in serum as diagnostic biomarker for Alzheimer's disease. J Proteome 75:3617–3629
Yin H, Kuret J (2006) C-terminal truncation modulates both nucleation and extension phases of tau fibrillization. FEBS Lett 580:211–215
Yuzwa SA, Shan X, Jones BA, Zhao G, Woodward ML, Li X, Zhu Y, McEachern EJ, Silverman MA, Watson NV, Gong CX, Vocadlo DJ (2014) Pharmacological inhibition of O-GlcNAcase (OGA) prevents cognitive decline and amyloid plaque formation in bigenic tau/APP mutant mice. Mol Neurodegener 9:42
Zhang Z, Song M, Liu X, Kang SS, Kwon IS, Duong DM, Seyfried NT, Hu WT, Liu Z, Wang JZ, Cheng L, Sun YE, Yu SP, Levey AI, Ye K (2014) Cleavage of tau by asparagine endopeptidase mediates the neurofibrillary pathology in Alzheimer's disease. Nat Med 20:1254–1262
Zimova I, Brezovakova V, Hromadka T, Weisova P, Cubinkova V, Valachova B, Filipcik P, Jadhav S, Smolek T, Novak M, Zilka N (2016) Human Truncated Tau Induces Mature Neurofibrillary Pathology in a Mouse Model of Human Tauopathy. J Alzheim Dis 54:831–843
Acknowledgement:
We thank the Eurotau organizers for an excellent meeting (http://www.lucbuee.fr/crbst_10.html). IG is supported by the following grants, ISF 1424/14, ERA-NET neuron AUTYSM, AMN Foundation as well as Drs. Ronith and Armand Stemmer and Mr Arthur Gerbi (French Friends of Tel Aviv University), and Canadian and Spanish Friends of Tel Aviv University. KH is supported by Hjärnfonden (FO2017-004), Aina Wallströms & Mary-Ann Sjöbloms Foundation, Ulla & Gerhard Hobohms Foundation.
Author information
Authors and Affiliations
Corresponding author
Ethics declarations
Competing Interests
IG is the Chief Scientific Officer of Coronis Neurosciences (www.coronisns.com).
Rights and permissions
About this article
Cite this article
Gozes, I., Höglinger, G., Quinn, J.P. et al. Tau Diagnostics and Clinical Studies. J Mol Neurosci 63, 123–130 (2017). https://doi.org/10.1007/s12031-017-0983-0
Published:
Issue Date:
DOI: https://doi.org/10.1007/s12031-017-0983-0