Download as pdf or txt
Download as pdf or txt
You are on page 1of 22

Journal of Alzheimer’s Disease 61 (2018) 487–508 487

DOI 10.3233/JAD-170187
IOS Press

Review

Tauopathies: Mechanisms and Therapeutic


Strategies
Chen-Chen Tan1 , Xiao-Yan Zhang1 , Lan Tan∗ and Jin-Tai Yu∗
Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University,
Qingdao, Shandong, China

Accepted 15 September 2017

Abstract. Tauopathies are morphologically, biochemically, and clinically heterogeneous neurodegenerative diseases defined
by the accumulation of abnormal tau proteins in the brain. There is no effective method to prevent and reverse the tauopathies,
but this gloomy picture has been changed by recent research advances. Evidences from genetic studies, experimental animal
models, and molecular and cell biology have shed light on the main mechanisms of the diseases. The development of
radiology and biochemistry, especially the development of PET imaging, will provide important biomarkers for the clinical
diagnosis and treatment. Given the central role of tau in tauopathies, many treatments have constantly emerged, including
targeting phosphorylation, targeting aggregation, increasing microtubule stabilization, tau immunization, clearance of tau,
anti-inflammatory treatment, and other therapeutics. There is still a long way to go before we obtain drug therapy targeted at
multifactor mechanisms.

Keywords: Biomarkers, mechanisms, neurodegenerative diseases, tau, tauopathy

INTRODUCTION integrate and interpret recent advances that have pro-


vided new clues for mechanisms and therapeutics of
Tauopathies are believed to be involved in dozens tauopathies. Moreover, we also discuss the promising
of neurodegenerative diseases, clinically manifesting and novel therapeutic strategies for other neurode-
with a wide spectrum of symptoms, including cog- generative disorders with the lessons learned from
nitive, behavioral, or motor impairments [1]. They the recent knowledge of tauopathies.
are histopathologically defined by the filamentous
inclusions of unusually phosphorylated tau protein TAU ISOFORMS AND GENETICS
in neurons and neuroglia [2]. Recently, mechanisms
and therapeutic strategies of tauopathies become a Tau is a member of microtubule-associated pro-
hot spot in this field. The discoveries of biomarkers tein family which is concentrated in axons of central
of tauopathies open up a new avenue for revealing nervous system and peripheral nervous system, and
the real roles of tau in mechanisms of neurode- plays a physiological role in dendrites [4]. Located
generative diseases and provide effective evidences on chromosome 17q21, a single gene containing 16
for clinical treatments [3]. In this review, we will exons (E) encodes the human tau proteins. Six tau iso-
1 Theseauthors contributed equally to this work. forms have been formed by alternative splicing of E2,
∗ Correspondence to: Lan Tan and Jin-Tai Yu, Department of 3, and 10 (Fig. 1A). The alternative splicing of E2 and
Neurology, Qingdao Municipal Hospital, School of Medicine,
Qingdao University, No. 5 Donghai Middle Road, Qingdao,
3 produces three tau isoforms-two N-terminal inserts,
Shandong Province 266071, China. E-mails: dr.tanlan@163.com one insert or no insert, while the alternative splicing
(Lan Tan); yu-jintai@163.com (Jin-Tai Yu). of E10 generates two tau isoforms-3R (three repeat

ISSN 1387-2877/18/$35.00 © 2018 – IOS Press and the authors. All rights reserved
488 C.-C. Tan et al. / Tauopathies: Mechanisms and Therapeutic Strategies

identified to date [4] (Fig. 1B). Mutations in E9-12


increase the mRNA splicing, leading to tau hyper-
phosphorylation, tangle formation, synapse loss, glial
activation, neuronal loss, and memory impairment
[7–9]. Missense mutations of MAPT jeopardize phys-
iological functions of tau protein and facilitate the
aggregation of microtubule. Nearly 80% of mis-
sense mutations are associated with frontotemporal
dementia and other neurodegenerations including
progressive supranuclear palsy (PSP), Pick’s dis-
ease (PID), and corticobasal degeneration (CBD)
[10]. Mutations of R406W and A152T are found
to be associated with both FTDP-17 and AD-like
pathological process [11, 12]. A152T increases the
levels of p-tau protein and decreases clearance of
p-tau protein. A152T substitution increases the risk
of neurodegeneration by increasing p-tau levels and
enhancing network hyperexcitability, combined with
other pathogenic factors [13]. Interestingly, the same
mutation was linked with distinct syndromes in the
same family; for example, the son has the clinical
manifestation of CBS while the father has frontotem-
poral dementia (FTD) [4].
Two haplotypes, including H1 with a 900-kb
Fig. 1. Tau six isoforms and mutation of MAPT in human brain.
A) Six tau isoforms are produced via inserting a 29-aminoacid inversion polymorphism and H2 with non-inversion
or 58-aminoacid encoded by E2 (light brown) and E3 (brownish polymorphism, also contribute to the neurodegenera-
yellow) in the N-terminal half and by the presence or absence of tive diseases. H1 causes PSP and Parkinson’s disease
31-aminoacid repeat domains encoded by E10 (brown) in carboxy-
terminal half. The alternative splicing E2 and E3 produces three
as well as CBD through H1c. H1c is located in one of
tau isoforms-2N (two N-terminal inserts), 1N (one insert) or 0N intron 0 regulatory region of MAPT. Recent studies
(no insert). The alternative splicing of E10 generates two tau have shown that MAPT H1c haplotype (rs242557)
isoforms-3R (three microtubule-binding repeat domains) or 4R is a genetic risk factor for CBD and PSP [14]. H2
(four microtubule-binding repeat domains). E1, 4, 5, 7, 9, 11,
12, and 13 (grey) constitute the basic component of tau protein,
confers protection by increasing E3 expression of
whereas E0, 4a, 6, 8, and 14 are not. B) Mutations of MAPT genes, MAPT in grey matter [15]. KANSL1 is a gene encod-
mainly relation with frontotemporal dementia and parkinsonism ing chromatin modifier which affects the expression
linked to chromosome 17. 55 sites mutations in MAPT including of gene via the lysine 16 acetylation of histone H4.
43 exon mutations and 15 non-coding-region mutations flanking
E10 are revealed. MAPT, microtubule-associated protein tau. Its haploinsufficiency causes 17q21.31 microdeletion
syndrome [16]. Additionally, the mutations in 18q
deletion syndrome and other 21 genes have been
domains) or 4R (four repeat domains) [5]. Except reported to be related to tauopathies. In the other
promoting the formation of microtubules and keeping 21 genes, some genes (ATP6AP2, SLC9A6, LRRK2,
their stability, tau also plays a key role in maintain- PRKN, SNCA, and CYP27A1) were detected to occur
ing neuronal architecture and axonal transport and with tauopathy through some “specific” mutations.
serves as an anchor for enzymes of cell signaling or Others (including CLN6, CNBP, DMPK, ITM2B,
other proteins. Meanwhile, it regulates cell activity NPC1, NPC2, PANK2, PRNP, PSEN2 and SLC17A5)
and viability [6]. were detected to form AD-like neurofibrillary tangles
A great majority of genetic studies put focus on (NFTs) in tauopathy (Table 1) [17].
the mutation of MAPT and related genes. Mutation
of MAPT and related genes promote tau aggregation, CLINICOPATHOLOGICAL FEATURES
reduce its affinity for microtubule, disturb the bal- OF THE TAUOPATHIES
ance between 3R and 4R and regulate the splicing of
mRNA [7], leading to tauopathies. Over 50 mutations Tauopathies are morphologically, biochemically,
on MAPT, which is located on 17q21-22, have been and clinically heterogeneous neurodegeneration
C.-C. Tan et al. / Tauopathies: Mechanisms and Therapeutic Strategies 489

Table 1
Genetic mutation with tau pathology
Diseases Mutation associated with tau
Alzheimer’s disease (AD) haplotypes H1/H2
Argyrophilic grain disease (AGD) MAPT (S305S, S305I)
Corticobasal degeneration (CBD) MAPT (N296H, N296N, P301L, P301S, S305N, G389R, R406W, N410H,
E10 + 16), H1 haplotype
Frontotemporal dementia and parkinsonism linked MAPT (A152T, K257T, L266V, G272V, N279K, 280K, N296, K298E,
to chromosome 17(FTDP-17) P301L, P301S, S305N, S305S, K317M, G335S, V337M, G389R, R406W,
E10 + 3, + 12, + 14, + 16) or progranulin
Frontotemporal lobar degenerations (FTLD) MAPT (G55R, I260V, L266V, G272V, N296H, P301S, P301T, L315R, S320F,
P332S, E342V, S356T, V363A, V363I, T427M)
Pick’s disease (PiD) MAPT (K257T, G272V, N279K, S305N, S320F, K369I, G389R)
Progressive supranuclear palsy (PSP) MAPT (R5L, L284R, P301L, S305S, E10 + 14, + 16), H1 haplotype
Guadeloupean parkinsonism H1 haplotype
Hemimegalencephaly (HME) AKT1, AKT3
Neurodegeneration with brain iron accumulation PLA2G6
Prion protein cerebral amyloid angiopathy Codon 145 of PRNP
Psychotic Alzheimer’ s disease H2 allele of the extended MAPT haplotype
Creutzfeldt-Jakob disease (CJD) PRNP
Familial British dementia (FBD) ITM2B
Familial Danish dementia (FDD) ITM2B
Niemann-Pick disease, type C NPC1, NPC2

defined by the accumulation of abnormal tau pro- [21]. Clinically, AD presents with typical and atyp-
teins in the brain. The neuropathological phenotypes ical manifestations including memory and learning
distinguish themselves from each other according impairment, executive dysfunction, aphasia, visual,
to different tau morphological features (pretangles, practical problems, and mental symptoms in the late
NFTs, threads, Pick bodies, dystrophic neuritis, of AD [22].
grains, and spherical cytoplasmic inclusions) and tau
isoforms (3R and 4R) [18]. Primary age-related tauopathy
Primary age-related tauopathy (PART) replaces
3R and 4R tauopathies these names (“tangle-predominant senile dementia”,
“tangle-only dementia”, “preferential development
Alzheimer’s disease of NFT without senile plaques”, and “senile demen-
Alzheimer’s disease (AD) is the most prevalent tia of the neurofibrillary tangle type”) to describe the
neurodegenerative disease associated with age- neurofibrillary tangle pathology that is observed in
related dementia. It is characterized by both A␤ the brains of aged individuals [23]. In comparison
extracellular plaques and intracellular neurofibril- with AD, PART has an association with MAPT tau
lary tangles made up of 3R and 4R tau [19, 20]. H1 haplotype while having no genetic risk associated
Pretangles are cytoplasmic tau immunoreactivity in with apolipoprotein gene (␧4) [24]. However, neu-
neurons without apparent formation of fibrillary rofibrillary tangles in PART are made up of 3R and
structures. In Alzheimer’s disease, such tau depo- 4R tau as those in AD. Hence, it is difficult to dis-
sition is considered to represent a premature state tinguish AD and PART. The relation between PART
prior to fibril formation (AD-pretangles), later to form and AD is still not clear.
neurofibrillary tangles and finally ghost tangles. This
morphological evolution from pretangles to ghost Chronic traumatic encephalopathy
tangles is parallel with their profile shift from four Chronic traumatic encephalopathy (CTE) is a
repeat (4R) tau-positive pretangles to three repeat progressive neurodegenerative disease caused by
(3R) tau-positive ghost tangles with both positive repetitive head injuries [25]. The symptoms will
neurofibrillary tangles in between. This complemen- be present after a long period of latency. The
tary shift of tau profile from 4R to 3R suggests that periods for different individuals vary from sev-
these tau epitopes are represented interchangeably eral years to several decades. It classically presents
along tangle evolution. The intracellular neurofibril- with mood, cognitive, and behavioral symptoms,
lary tangles formed by abnormal tau proteins cause including depression, aggression, impulsivity, suici-
deficits through a loss-of-function mechanism in AD dal ideation, apathy, short-term memory loss, and
490 C.-C. Tan et al. / Tauopathies: Mechanisms and Therapeutic Strategies

executive dysfunction [26]. The concept was first akinesia with gait freezing (PAGF), PSP-bvFTD, and
introduced in 1927 [26], and the symptoms of CTE PNFA [35]. The cortical tau pathology was more
was reported by Harrison Martland in 1928 [27]. significant in PSP-CBS, PSP-bvFTD, and PNFA
Subsequently, hyperphosphorylated tau was found in than in PSP-P and PAGF. PSP-P and PAGF are
small cerebral vessels, cerebral sulci, periventricu- brainstem-predominant PSP syndromes. Compared
lar areas, subpial regions, subcortical structures, and with PSP-RS, they showed more severe degeneration
brainstem nuclei and it also had an immune response and less cortical tau pathology in substantia nigra,
for both 3R and 4R tau [26]. Besides, the astrocytic globus pallidus, and subthalamic nucleus [36].
tau pathology was observed in periventricular and
subpial areas, and astrocytic tangles were formed in Argyrophilic grain disease
CTE [25]. Argyrophilic grain disease (AGD) is described
in amygdale, entorhinal cortex, hippocampus, and
4R tauopathies neighboring temporal cortex in patients with adult
onset dementia and is characterized by spindle-
Corticobasal degeneration shaped “grains” [37]. In peri-amygdaloid white
Corticobasal degeneration (CBD) is related with matter and hippocampus, AGD presents with oligo-
huge burden of tau inclusions in white matter, basal dendroglial coiled bodies and pretangles [18].
ganglia and brainstem. It classically presents with Besides, tau pathology was found in different
motor features (myoclonus, dystonia, gait abnor- regions in different stages. The changes in AGD
malities, bradykinesia, limb and axial rigidity) and are dependent on tau neurofibrillary tangles, which
higher cortical features (cortical sensory loss, apha- are biochemically difficult to distinguish from AD.
sia, apraxia, cognitive impairment, alien limb, and Cognitive decline, episodic memory loss, demen-
behavioral changes) [28]. Some patients showed a tia, personality changes, aphasia, emotional and
frontotemporal pattern of cognitive deficits (such as mood imbalance (irritability, delusions, amnesia,
personality change, a progressive non-fluent aphasia apathy, irritability, dysphoria and agitation), promi-
and impaired awareness of deficit) and supranuclear nent abnormal behavior, and aggression have been
gaze palsy [29]. Besides these clinical symptoms, noted in the patients of AGD, but no specific clinical
behavioral changes and psychiatric symptoms with- syndrome was found to diagnose this disease [38].
out motor symptoms were also found in CBD, but
less severe degenerative changes were found in the 3R tauopathies
substantia nigra and subthalamic nucleus of these
patients [30]. CBS, CBD-bvFTD, PSP syndrome, and Pick’s disease
progressive non-fluent aphasia (PNFA) were 4 clin- PID is first used to describe atrophy of the
ical subtypes of CBD [31]. Patients with CBD and frontotemporal lobes in patients with behavioral dis-
Richardson syndrome have greater atrophy in anterior turbances and progressive language disorder, now
corpus callosum, more severe neuronal loss in sub- used to describe the disease with tau-positive intra-
stantia nigra and less neuronal loss in the subthalamic neuronal “Pick body” inclusions [39]. Clinically, it
nucleus, when compared with PSP [32]. presents with the disorder of social comportment,
executive impairments, PPA, visuospatial deficits
Progressive supranuclear palsy and akinetic-rigid syndrome with executive. The
PSP refers to Richardson’s syndrome or PSP clinical symptoms of PID were associated with
syndrome [33]. In 1964, Richardson’s syndrome bvFTD, PPA, and CBS [40]. The autopsy found other
(PSP-RS) was first described by Steele, Richardson, tauopathies including CBD, PSP, AGD, AD and TDP-
and Olszewski [34]. Early postural instability/falls, 43 proteinopathies in bvFTD patients [41]. The poor
supranuclear gaze palsy, variable cognitive impair- specificity of bvFTD for PID neuropathology neces-
ments, apraxia of speech and pseudobulbar affect sitated the recent clinical research criteria for bvFTD
were also observed in patients [19]. Abnormal accu- in order to more accurately predict PID. The stud-
mulation of 4R-tau proteins was seen in PSP patients, ies of PPA variants showed various results. Semantic
and they were present in different neuroanatomical variant PPA (svPPA) and PNFA were present in dif-
patterns to produce variable clinical features [33]. ferent tauopathies. The former is due to TDP-43
PSP variants are well-recognized in recent years and proteinopathies and the latter is due to PSP and CBD.
include PSP-parkinsonism (PSP-P), PSP-CBS, pure The “Pick body” inclusions were mainly made up of
C.-C. Tan et al. / Tauopathies: Mechanisms and Therapeutic Strategies 491

3R tau and distributed in hippocampus, dentate gyrus, Biomarkers of tau in neuroimaging


and cortical areas [42]. The pathogenesis of muta-
tions in tau proteins has been mentioned in FTDP-17. Positron emission tomography (PET) may be one
Recent studies have developed a novel 3R mutant tau of the noninvasive imaging technologies to measure
transgenic model for better understanding the natural tauopathies and associated neurodegeneration in
history, progression and therapy of PiD [43]. More- the brain. PET imaging of tau pathology relies on
over, a novel missense mutation in E12 of MAPT two major radioactive tracers: 18 F-labeled (t1/2 =
(p.Q336H) has been suggested to cause PiD, possibly 109.8 min) tracers and 11 C-labeled (t1/2 = 20.3 min)
by increasing 3R tau isoforms aggregation [44]. tracers [45]. With the development of the PET trac-
ers, many tracers were reported to be applied to the
BIOMARKERS OF TAUOPATHIES detection of tau pathology including 18 F-FDDNP,
18 F-THK523, 18 F-THK5117, 18 F-THK5105,
18 F-THK5351, 18 F-AV-1451(T807), 18 F-T808, 11 C-
A biomarker is a parameter which reflects the
progress of disease and the effects of treatment. PBB3, 11 C-THK951, lansoprazole, RO6931643,
Recently, neuroimaging biomarkers of tauopathies RO6924963, and RO6958948 [3, 46–52]. The char-
will provide strong underpinnings for prevention, acteristics of these tracers were described in Table 2.
diagnosis, and therapeutic targeting. A phase 0 trial of 18 F-FDDNP to evaluate tau values

Table 2
Tracers of PET imaging for tau
Tracers Affinity to tau Characteristics Reference
18 F-FDDNP Nanomolar affinity Non-selectivity for intercellular NFTs and extracellular [3]
A␤ plaques
18 F-THK523 High binding affinity and 12-fold Lower selectivity to PHF tau, in gray matter than white [46]
selectivity for tau than A␤ matter, not detected in non-AD tauopathies and
asynuclein deposits
18 F-THK5105 Higher binding affinity than 18 F-THK523 Relation with brain atrophy and dementia severity; a [47, 48]
relatively slower clearance from the brain and higher
lipophilicity; a lower signal to noise ratio
18 F-THK5117 Higher binding affinity than 18 F-THK523 Relation with brain atrophy and dementia severity [49]
Higher pharmacokinetics and better signal-to-noise
ratios than 18 F-THK5105
18 F-THK5351 high tau binding affinity in AD brains Higher signal-to-noise ratios, faster kinetic and lower [3]
white matter retention than 18 F-THK5105
and18 F-THK5117
18 F-AV-1451(T807) Higher nanomolar affinity and 25-fold Associated with diseases severity, distribution of [50]
selectivity for PHF tau over A␤ retention coincides with PHF tau and lower retention
in white matter; rapid washout, brain extraction and
no plasma metabolites entering the brain
18 F-T808 Higher nanomolar affinity and 25-fold The same function with18 F-T807 more rapid distribution [50]
selectivity for PHF tau over A␤ in the brain and maintain a steady state during the
scan, substantial defluorination phenomenon
11 C-PBB3 a 40–50 times higher affinity for NFTs Not only present in AD but in CBD and PSP and [49]
than for senile plaques candidate tracers were for tau but not A␤ and crossed
the blood–brain barrier
Lansoprazole Subnanomolar affinity to tau fibrils No entry into the brain but labeled with 11 C or18 F will [51]
entry into the brain without obstacles
11 C -THK951 slightly lower affinity to tau low lipophilicity, fast clearance and rapid brain uptake [52]
RO6931643, high-affinity binders on tau aggregates at Rapid brain entry, washout and safe metabolic patterns [52]
RO6924963 and the 3 H-T808 binding site and lack
RO6958948 affinity to A␤
18 F-FDDNP: 2-(1-(6-[(2-[18 F] fluoroethyl) (methyl) amino]-2- naphthyl) ethylidene) malononitrile; 18 F-THK523:2-(4-aminophenyl)-
6-(2-[18 F] fluoroethoxy) quinoline; 18 F-THK5105:6-[(3-[18 F] fluoro-2-hydroxy) propoxy]-2-(4-dimethylaminophenyl) quinolone;
18 F-THK5351:6-[(3-18 F -2-hydroxy) propoxy]-2-(4-methylaminopyridyl) quinoline; 18 F-T807: (E)-4-(2-(6-(2-(2-(2-[18 F] fluoroethoxy)

ethoxy) ethoxy) pyridin-3-yl) vinyl)-N-methyl benzenamine; 18 F-T808:2-(4-(2-[18 F] fluoroethyl) piperidin-1-yl) benzo (4, 5) imidazo (1, 2-a)
pyrimidine; 11 C-PBB3: a tau PET tracer constructed around a phenyl/pyridinyl-butadienyl-benzothiazole/ benzothiazolium (PBB) scaffold;
NFTs, neurofibrillary tangles; PHF, paired helical filaments; AD, Alzheimer’s disease; CBD, Corticobasal degeneration; PSP, Progressive
supranuclear palsy.
492 C.-C. Tan et al. / Tauopathies: Mechanisms and Therapeutic Strategies

in Parkinson’s disease dementia has been completed, impairment patients and cognitively healthy subjects
but no results have been posted (https://clinicaltrials. influenced by heterogeneous neurological disorders
gov/ct2/show/NCT02243982). 13 clinical trials for with a higher sensitivity of 85% and a low specificity
18 F-AV-1451 have been recruiting patients. Two trials of 65% [59]. It is not a biomarker for the diagnosis
of 18 F-AV-1451 have been completed, but no results of cognitive impairment.
are reported (https://clinicaltrials.gov/ct2/show/
NCT02079766 and https://clinicaltrials.gov/ct2/ Biomarkers of tau in peripheral blood
show/NCT01992380). In the living human brain,
18 F-AV1451 makes it possible for the regional Platelets are not only the main reserve of the
distribution of tau pathology visually [53]. A phase amyloid precursor protein, but include several tau
II trial for18 F-AV1451 to measure the distribution of molecules. High molecular weight tau proteins were
tau proteins in living human brain is ongoing (https:// detected in platelets of peripheral blood in AD
clinicaltrials.gov/ct2/show/NCT02191267). At the [60]. Tau fragments, tau-A and tau-C (a caspase-
same time, 18 F-T807 is also in phase II process 3 cleaved tau fragment) are detected in serum,
as a diagnostic PET imaging tracer for AD and which serve as the novel biomarker in serum for
neurodegeneration characterized by tau pathology the neurodegeneration [61–63]. Recent studies have
[54]. Besides, 18 FMNI-798 was used to evaluate shown that plasma tau is not an AD biomarker
tau protein burden in AD in a phase I and II trial in individual people, and it just partly reflects AD
(https://clinicaltrials.gov/ct2/show/NCT02640092]). pathology [64]. The levels of tau proteins were sig-
RO6931643, RO6924963, and RO6958948 as the nificantly increased in serumas indicators for severe
new tracers for PET are being tested in humans, and TBI and Olympic boxing [65]. Minor axonal dam-
a phase I trial has been completed to assess the ability age induced by punches causes the increase of
to detect tauopathy (https://www.clinicaltrials.gov/ plasma tau. The plasma tau will significantly increase
ct2/show/NCT02187627). after a bout in 1–6 days [65]. Circulating miR-
NAs also provide the evidence for AD. The serum
Biomarkers of tau in cerebrospinal fluid levels of cell-free miR-125b in serum decrease in
AD [66].
CSF tau species have been considered as the poten-
tial biomarkers for tauopathies, which are useful to MECHANISMS OF TAUOPATHIES
diagnose and identify different forms of neurodegen-
eration. Several researchers found that CSF t-tau and Tau hyperphosphorylation
p-tau have been considered as biomarkers for AD, and
the concentration of t-tau in AD has a two- to three- Phosphorylation plays the pivotal role in phys-
fold increase than in controls, while the p-tau in CSF iological and pathological function of tau. In the
shows a specificity of 80–100% [55]. Tau phospho- longest tau isoform, there are 85 potential sites of
rylation sites found in CSF up to now are Ser199, phosphorylation–45 serines, 35 threonines and 5
Thr231, Thr 181, Ser 396/404, and Ser235 [55]. As tyrosines [67]. Among them almost 40 sites become
for p-tau181, it has low specificity and sensitivity to abnormal phosphorylated in tauopathies [68]. Phos-
distinguish AD and non-AD cases [56], and this abil- phorylation sites in tau are in the C-terminal tail
ity increases after binding to A␤1-42 and t-tau [57]. region and proline-rich region [69]. Many factors
The diagnostic power of A␤1-42 /p-tau181 is higher such as accelerated aging, traumatic brain injury
than A␤1-42 and t-tau in the discrimination between (TBI), LRRK2, hypothermia, ␤-N-methylamino-
AD and FTD. When focusing on the discrimination L-alanine (BMAA), ischemia, oxidative stress,
between AD and CJD, p-tau181P/T-tau shows signif- methanol, and Cytoplasmic FMR1 interacting pro-
icantly higher diagnostic power than A␤1-42 , t-tau, tein 2 (CYFIP2) induce hyperphosphorylation of tau
and A␤1-42 /p-tau181P. [70–75]. Among them, accelerated aging contributes
Besides AD, a reduced ratio of CSF p/t-tau has to an acceleration of tau hyperphosphorylation,
been proved to have 82.4% positive predictive value leading to the development of behavioral changes
to detect TDP pathology in FTLD [58]. The levels which influence tau pathogenesis [75]. Recent stud-
of the 20–22 kDa NH2-truncated form of human tau ies indicated that tau phosphorylation is a risk factor
in CSF could be rarely discovered in non-demented associated with the extent of cognitive impairment
cases, which could distinguish between cognitive due to TBI. Meanwhile, elevation of GSK-3␤ kinase
C.-C. Tan et al. / Tauopathies: Mechanisms and Therapeutic Strategies 493

activity and reduction in PP2A phosphatase activ- Tau aggregation


ity have been suggested as, respectively, positive and
negative factors associated with the severity of TBI. Tau is a highly soluble unfolded protein and not
Altogether, these results suggest a role of tau phos- aggregated with each other in physiological con-
phorylation pathway for the alterations of injured ditions and concentrations. Tau aggregation is not
neurons in mediating cognitive damages of TBI [76]. only the result of the phosphorylation, but also can
Tau phosphorylation is exquisitely sensitive to tem- be affected by other factors. Negatively charged
perature, increasing by 80% for each degree below molecules are the inducers of non-phosphorylated tau
37◦ C, due to exponential decrease in PP2A activ- aggregation [4], including heparin, anionic micelles,
ity during direct hypothermia, or anesthesia-induced RNA, synthetic particles, fatty acids, fatty acid-
hypothermia [77]. BMAA leads to tau hyperphos- like molecules (such as arachidonic acid), and lipid
phorylation by activating metabotropic glutamate vesicles. Heparin induces the polymerization of
receptor 5 (mGluR5). Then, mGluR5 results in tau fragments by binding to the second and third
the release of PP2Ac and its phosphorylation at microtubule-binding repeats [85], while arachidonic
Tyr307 to inhibit PP2A [71]. Transient brain ischemia acid is more effective than heparin in promoting
has been shown to induce tau hyperphosphoryla- aggregation of full-length tau when the number
tion. Recent studies confirmed that tau protein was of K18, a tau fragment identified as one of the
dephosphorylated during brain ischemia. In addition, four repeats of MBD, which is united to the
the activity of GSK-3␤ increased and the activity lipid bilayer, oversteps a critical surface density
of PP2A decreased. After reperfusion, tau protein [86, 87].
was hyperphosphorylated; the activity of GSK-3␤ Acetylation of tau prevents degradation of phos-
decreased; the activity of PP2A remained low. Impor- phorylated tau, leading to tau aggregation [88]. In an
tantly, the interaction of tau with GSK-3␤ and animal model of tauopathy, the insoluble tau frac-
PP2A was altered during ischemia and reperfusion tion was observed after the acetylation, suggesting
[78, 79]. Methanol not only leads to acute neuro- that acetylation promoted soluble tau to insoluble
logical sequelae, including blindness and death, but tau in order to affect the process of aggregation
low concentrations of methanol also lead to hyper- [89]. CREB binding protein (CBP) and p300 acety-
phosphorylation of tau and apoptosis of hippocampus late different residues of tau, changing tau’s intrinsic
neurons [74]. The lack of CYFIP2 increases the propensity to aggregate [90]. In addition to acetyl-
expression of alpha-calcium/calmodulin-dependent transferase, acetylation also regulates the process of
kinase II in order to exacerbate tau hyperphospho- tau aggregation through other ways. At KXGS motifs,
rylation [80]. acetylation and phosphorylation act in a competi-
MicroRNAs (miRNAs) participate in the progress tive relationship and are regulated by HDAC6 [90].
of tau phosphorylation, such as miR-922, miR-125b, The inhibitors of HDAC6 facilitate tau acetylation
and miR-132. MiR-922 induces tau phosphoryla- and prevent tau phosphorylation at these motifs to
tion through reducing the expression of ubiquitin interfere with tau aggregation propensity. In addi-
carboxy-terminal hydrolase L1 (UCHL1), and affects tion, HDAC6 could also impact on turnover of tau
tau phosphorylation through influencing the num- through regulating the acetylation of its substrate
bers of NFTS [81]. MiR-125b promotes tau Hsp90, suggesting combination treatment of HDAC6
hyperphosphorylation by its overexpression. When and Hsp90 inhibitors as a potent therapeutic treatment
miR-125b was injected into mice, cdk5, p35, and for diseases with accumulation of tau [91]. Recently,
p44/42-MAPK signaling were upregulated and the ac-K174 has a critical role in tau accumulation and
anti-apoptotic factor Bcl-W and the phosphatases toxicity. Besides, the acetyl-mimic mutant K174Q
DUSP6 and PPP1CA were suppressed, leading to inhibits tau degradation and promotes tau aggrega-
tau phosphorylation [82]. MiR-132 loss promotes tion [92]. Recent studies suggest that stress granules
tau pathology through regulating the expression of (SG) are a form of pathology associated with neu-
inositol 1,4,5-trisphosphate 3-kinase B (ITPKB), rodegenerative diseases. SG proteins, such as TIA-1
which regulates tau phosphorylation [83]. In the and TTP, bind to phosphorylated tau, and that binding
Drosophila tauopathy model, proto-oncogenes sup- increases with the severity of the disease. Meanwhile,
presses tau-mediated neurodegenerative diseases SG proteins might contribute to generating tau pathol-
by reducing abnormal tau hyperphosphorylation ogy, perhaps by creating local environments with high
[84]. concentrations of phospho-tau [93]. Latest studies
494 C.-C. Tan et al. / Tauopathies: Mechanisms and Therapeutic Strategies

suggest that the pathological tau aggregation may Mechanism of autophagy-lysosome system
be initiated through the regulated self-assembly of in tauopathies
core SG-associated RNA binding proteins, such as
TIA-1 [94]. Autophagy is a catabolic system which can degrade
Additionally, truncation at both ends (N-terminal abnormal proteins, lipids and organelles through
or C-terminal) of the tau molecule could confer a lysosomes. It plays an important role in cell home-
toxic gain of toxic function. The in vivo model of ostasis and keeps the metabolic balance between
tauopathy based on the truncated tau protein clearly synthesis and degradation and subsequent turnover
shows that truncation is a “productive” modifica- of cytoplasmic materials in a stressful environment.
tion that can initiate tau neurofibrillary degeneration Autophagy is classified into three types: microau-
[95]. The truncated forms of tau protein are espe- tophagy, chaperone-mediated autophagy (CMA) and
cially found in PHFs, suggesting that tau truncation macroautophagy [103, 104]. Many factors inhibit
might contribute to tau aggregation [96]. Recent stud- autophagy which leads to tau accumulation in neu-
ies suggest that there is a complex interaction between rodegenerative diseases, including mTOR, p53 tumor
phosphorylated and truncated tau species [97]. More- suppressor, cAMP, NH4Cl, cathepsin inhibitors,
over, tau truncation could modulate tau spreading. 3-methyladenine, chloroquine, Bcl-2, Bcl-XL, and
Finally, changes in tau 3R/4R ratio may result in increased concentration of Ca2+ in cytoplasm which
differences in microtubule stability. comes from the endoplasmic reticulum [105–108].
The mTOR is a primordial inhibitory signal which
Neurotoxicity of tau oligomers participates in the initial process of signal transduc-
tion. When growth factors such as insulin-like growth
As mentioned above, hyperphosphorylation of tau factor bind to insulin-like growth factor receptors
consists of neurofibrillary tangles (NFT). Before the (IGF1R), class I PT3K pathway, which catalyzes the
formation of NFT, tau forms two intermediate forms conversion of PIP2 to PIP3, is activated. PIP3 recruits
of NFT: tau oligomers, not discovered by atomic AKt and DDK1 to membrane, allowing the latter one
force microscopy (AFM), and granular tau oligomers, to phosphorylate AKt. The active AKt inhibits the
detected by AFM [1]. Tau oligomers play a cru- activity of the TSC1/TSC2 complex, whose activ-
cial role in neurodegeneration, synaptic dysfunction ity can lead to an overall inhibition of mTORC1
and mitochondrial loss. Tau oligomers impair mem- signaling. By contrast, intracellular signals include
ory by disrupting anterograde memory storage when nutrient starvation, low energy levels (increase of the
researchers inject the monomers of full-length recom- AMP/ATP ratio), hypoxia, and DNA damage inhibit
binant p-tau-441 or fibrils into the C57BL/6 wild mice mTOR activity [109]. CMA (chaperone-mediated
hippocampus [98]. Recent animal studies found that autophagy) may generate tau fragment aggregation
mice expressing TauC3 (a caspase-cleaved form of [110]. There are two CMA-targeting motifs in MBDs
tau) showed memory and learning impairment and of tau, 336QVEVK340 and 347KDRVQ351, which
the decrease of synaptic number at early stage accom- participate in the transportation of tau to lysosomal
panied with tau oligomer formation [99]. membranes. The overexpression of neuronal PAS
Tau oligomers cause synaptic dysfunction through domain protein 4 promotes the clearance of patholog-
affecting the density of presynaptic and neuronal traf- ical tau by inducing autophagy [111] and provides a
ficking and also cause mitochondrial dysfunction by novel therapeutic approach to tauopathies.
activating caspase-9 [98]. Intracellular p-tau accu-
mulation causes mitophagy deficits, which inserts Mechanism of ubiquitin-proteasome system
into the mitochondrial membrane to activate the in tauopathies
membrane potential and to cause the decrease
of PTEN-induced kinase 1/Parkin. PTEN-induced Like autophagy-lysosome system, the ubiquitin-
kinase 1/Parkin reduces p-tau-induced impairment proteasome system (UPS) is a primary mechanism
[100]. Except these, miR-219 targeting tau synthe- of organism that mediates the degradation of abnor-
sis directly causes the toxicity of tau [101]. Recently, mal or misfolded proteins and short-live proteins.
a new mechanism of tau toxicity is found to con- The system contains two parts–ubiquitination and
tribute to glial activity impairment in AD and other proteasome, which interact with each other to medi-
tauopathies, which induces microtubule bundling and ate the degradation of abnormal tau [112]. In the
activate the Rho-GTPase-ROCK pathway [102]. brain in AD, polyubiquitin chain of PHF-tau is
C.-C. Tan et al. / Tauopathies: Mechanisms and Therapeutic Strategies 495

found, which is attached to others through four lysine tau, released tau, and tau fragments via the pattern of
residues (Lys63, Lys48, Lys11, and Lys6) [106]. potassium-induced depolarization [121, 122].
The affinity between tau and microtubule is reduced
when tau is modified with ubiquitination. Meanwhile, Endocytosis spreading pathway
researchers found that E3 ubiquitin ligase activity Extracellular tau not only can aggregate tangles
played a role in the ubiquitination of tau [113]. E3 in interstitial fluid and CSF, but also can be inter-
ubiquitin ligase determines the specificity of interac- nalized via endocytosis. Uptake of tau fibrils occurs
tion between ubiquitin and substrates. through adsorptive endocytosis, which is time- and
Some inhibitors of proteasome, lactacystin, epox- temperature-dependent [123]. Another endocytosis
omicin, MG132, and leupeptin put the degradation may occur when extracellular tau interacts with
of abnormal tau off. TNF-␣/NF␬B reduces 26S receptors, which is activated by intracellular calcium
proteasome and then decreases the activity of pro- via M1/M3 muscarinic receptor stimulation [124].
teasome by S5b/PSMD5 pathway, which plays a Meanwhile, the interaction between tau and mus-
vital role in UPS-associated tauopathies [114]. Not carinic receptors generates toxic influences and leads
only in AD, but in transient cerebral ischemia, pro- to the release of tau into the extracellular space and
teasome dysfunction causes E10 exclusion of tau neuron death [125]. Other mechanisms have been
and a decrease of Tra2␤, leading to the imbalance identified, including the entry of non-fibrillar and sol-
of 4R/3R and neurodegeneration [115]. UBB + 1 uble tau aggregates via dynamin-driven endocytosis
interacted with polyubiquitin chains to inhibit the [126] and proteoglycan-mediated macropinocytosis.
degradation of polyubiquitinated substrate through Except abnormal tau, PBS-soluble phosphorylated
26S proteasome, a dose-dependent inhibitor of pro- high-molecular-weight tau takes part in tau prop-
teasome [116]. Maintained stability and activity of agation and is present in the form of endogenous
UPS is a potential therapy for neurodegenerative tau [127].
diseases and tauopathies.
Trans-synaptic spreading pathway
Propagation of tauopathies Synapses play a crucial role in the spreading
of tauopathy. Studies of human postmortem tissue
Preclinical phase of the disease, mild cognitive roughly described the anatomical distribution of the
impairment and severe dementia are three stages of tauopathies, and demonstrated that these pathology
clinical symptoms of AD. Each stage has two sequen- areas were linked by synapses [117]. Furthermore, in
tial neuropathological stages according to Braak vivo studies revealed that tau pathology was propa-
stages (stage I-II, stages III–IV, stages V–VI) and gated to other areas associated with synapses from
approximate three stages described by Delacourte the initial location [128]. How the precise process
(Delacourte stages 1–3, Delacourte stages 4–6 and of propagation happened is still uncertain, except
Delacourte stages 7–10) [3, 117]. The six or ten trans-synaptic and endocytosis spreading. Tau may
sequential neuropathological stages reveal the prop- be transferred through tunneling nanotubes between
agation of tau pathology, but the precise process of cells and membrane penetration [129]. Besides, new
tau propagation is still a mystery. research in animal models showed that microglia
and exosomes promoted tau propagation in the
Release of tau mammalian brain and revealed that microglia could
The appearance of extracellular tau tangles is iden- transfer tau to other neurons [130]. Recently, an
tified at stage III, and how tau is released is crucial animal model showed that amyloid promoted tau
for us to understand the process of extracellular tau propagation and increased the toxicity of tau [131].
tangle formation. Extracellular tau is not only from
the dying neurons but also from other unconven- POTENTIAL HYPOTHESIS MECHANISM
tional pathways [118]. Calcium affects the release of
endogenous tau by an unconventional pathway [119]. Although above-mentioned mechanisms can cause
However, the concentration of calcium also influ- tauopathies in their own ways, many evidences from
ences the process of endogenous tau secretion. The animal’s experiments, preclinical models and post-
release of tau is increased when the agonist S-AMPA mortem research show that the mechanisms interact
increases the activity of glutamate receptors [120]. with each other to cause tauopathy (Fig. 2). Abnor-
The pre-synaptic compartment consists of abundant mal tau proteins are mainly degraded through two
496 C.-C. Tan et al. / Tauopathies: Mechanisms and Therapeutic Strategies

Fig. 2. Mechanisms of tauopathies. In the brain, the mutations of genetic (①) and the disorder of splicing (②) induce the production
of abnormal tau proteins. Abnormal tau proteins detach from microtubule, leading to the instability of microtubule and the damage of
axon transport. Meanwhile, the imbalance of protein kinases and phosphatases and other factors can induce tau hyperphosphorylation (③).
Abnormal tau proteins and hyperphosphorylated tau aggregate with each other, non-phosphorylated tau also aggregate after interacting
with negatively charged molecules (④). The primary form of aggregation is tau oligomers and cause mitochondrial (⑤), then tau oligomers
polymerize to form NFTs (⑥) and cause tauopathies. The UPS (⑦) and autophagy-lysosome system (⑧), mediated abnormal tau degradation,
are blocked by the inhibitors, causing the increase of abnormal tau levels. Intracellular tau secretes to extracellular space via targeting to
special receptors (a), exosomes (b), and with the help of microglia (c). These abnormal tau pass to next neuron through endocytosis (f), trans-
synaptic pathway (d), or exosomes (e), resulting in new pathology. MTs, microtubules; NFTs, neurofibrillary tangles; DUBs, deubiquitinating
enzymes; 3MA, 3-methyladenine; CMA, chaperone-mediated autophagy; UPS, ubiquitin proteasome system.

systems–UPS and autophagy-lysosome system. Tau iments of treatment. Some crucial aspects of human
proteins induce neuron death and neurodegeneration tauopathies have been recapitulated in modified ani-
by a series of processes including hyperphosphoryla- mals including Caenorhabditis elegans, Drosophila,
tion, aggregation, oligomers, and then the formation and especially mouse models (Table 3) (see review in
of the NFTs. The NFTs are made up of paired helical [132]). Caenorhabditis elegans and Drosophila are
filaments (PHFs) and straight filaments, which are two easy tools for genetic studies. The Caenorhab-
composed of abnormal tau proteins. Subsequently, ditis elegans models have revealed tau toxicity,
abnormal tau in extracellular space propagates via axonopathy, motor dysfunction, and early death
endocytosis and trans-synaptic pathway and causes [133]. This model is also used to study drug screen-
the damage of neighboring neurons, leading to the ing [134]. The Drosophila models are used to identify
tauopathies. several pathological pathways or susceptibility genes.
Besides, these models profit from low redundancy of
EXPERIMENTAL ANIMAL MODELS the genome and low consumption [135].
OF TAUOPATHIES The first transgenic mouse models of human
tauopathies were produced by only expressing spe-
Experimental animal models of tauopathies serve cific human tau isoforms (4R/2N) [136]. These
as efficient systems for interpreting the role of models exhibited hyperphosphorylated tau and pre-
abnormal tau in the neurodegeneration and the exper- tangle tauopathy, and NFTs were not observed in
Table 3
The major transgenic mouse models of tauopathies
Mutation Promoters DNF initiation Histopathology Phenotype
G272V(4R2N) MoPrP ND Tau filament formation, transgene expression in oligodendrocytes and neurons No obvious neurological deficits
N279K(4R0N) MoPrP ND Phosphorylated tau in the olfactory bulb, hippocampus, cerebellum, Impairment in acquisition of spatial learning
diencephalons, brain stem and cerebral cortex and active avoidance, motor deficit and
cognitive decline
N279K(4R2N) hTau 13 months hence the 4R:3R ratio, tau protein accumulation in neurons and tufted astrocytes The degeneration of the nigrostriatal
dopaminergic pathway, motor and
behavioral deficits
P301L(4R0N) CaMK-II 1 months NFTs, neuronal loss Cognitive impairment and motor deficit
P301L(4R0N) MoPrP ND Neuronal loss, Age- and dose-dependent NFTs development, neurogenic muscle Motor and behavioral impairment, premature
atrophy death
P301L(4R0N) neuropsintTA 3 months Hyperphosphorylated tau, PHF1, insoluble tau aggregated, axonal degeneration ND
P301L(4R1N) 2 3 -CNP 3–9 months Impaired axonal transport, the disruption of myelin and axons preceding Progressive motor weakness and weight loss,
filamentous oligodendroglial tau inclusions probably a consequence of neurogenic
muscle atrophy
P301L(4R1N) GFAP 12–24 months Insoluble tau, age-dependent accumulation of tau inclusions in astrocytes Motor deficits and additional deficits in
neuromuscular strength
P301L(4R2N) mThy-1 6–7 months Tau hyperphosphorylation, widespread NFTs, no axonopathy Late minor motor phenotype, muscle and
tissue wasting, cognitive decline
P301L(4R2N) mThy-1.2 3 months NFT-like, insoluble, short filament structures with hyperphosphorylated tau gliosis Cognitive decline
P301L(4R2N) MoPrP 2–8 months age- and gene-dose-dependent development of NFT, insoluble tau, axonal motor and behavioral deficits
spheroids, anterior horn cell loss and axonal degeneration, Pick-body-like
neuronal lesions
P301S(4R0N) mThy-1.2 5–6 months Abundant filamentous tau in spinal cord and brain stem Motor deficits and cognitive decline,
behavioral abnormalities
P301S(4R1N) MoPrP 3–6 months Early synaptic pathology, neuronal loss, NFTs formation Motor deficits and cognitive decline, limb
weakness, progressing to paralysis
V337M(4R2N) PDGF␤ 11 months Neuronal cell death, PHF, degenerating hippocampal neurons Cognitive deficits, decreased hippocampal
neural activity
V337M(3R2N) mThy-1 12 months Insoluble tau, filamentous neurofibrillary tangles in the brain, aberrantly Cognitive decline
phosphorylated tau
R406W(4R2N) MoPrP 12 months Tau lesion with advancing age in neuronal perikarya, insoluble tau, tangles Motor deficits
C.-C. Tan et al. / Tauopathies: Mechanisms and Therapeutic Strategies

formation
R406W(4R2N) CaMK-II 14–18 months Hyperphosphorylated tau, insoluble tau filament in aged mice only, tangles Cognitive decline, no obvious sensorimotor
formation deficit
R406W(4R2N) Syrian hamster PrP 10 months Hyperphosphorylated, insoluble tau starting to aggregated in amygdala and Motor deficit and cognitive decline
hippocampus
G272V/P301S mThy-1.2 3–6 months NFTs, ghost tangles Behavioral abnormalities, cognitive decline
(4R1N)
G272V/P301L/ mThy-1 ND Insoluble filament and tangles formation ND
R406W(4R2N)
ND, not determined; Ref, reference; MoPrP, mouse prion protein promoter; CaMK-II, Ca 2 + /calmodulin kinase II; NFTs, neurofibrillary tangles; PHF1, paired helical filaments 1; 2 3 -CNP,
2 ,3 -cyclic nucleotide 3 -phosphodiesterase; GFAP, glial fibrillary acidic protein; PDGF␤, platelet-derived growth factor␤.
497
498 C.-C. Tan et al. / Tauopathies: Mechanisms and Therapeutic Strategies

these models. To observe the NFTs, novel models Targeting phosphorylation in tauopathies
were generated [132]. Meanwhile, the relationship
between neuronal loss and neurofibrillary pathology As a hallmark in AD and other neurodegenera-
was also found. Not only did the transgenic (tg) tive diseases, abnormal tau phosphorylation has the
mice models of P301L demonstrate the process of indelible effects on the process of pathology and
tauopathies in neuronal and glial, but G272V, N279K, diseases. The protein kinases provide a vital target
P301S, V337M, and R406W were created to explore site for tau phosphorylation, especially glycogen syn-
the function of tau in tauopathies [132]. These mod- thase kinase 3 (GSK-3). A number of GSK-3 kinase
els exhibited different aspects and initiation time of inhibitors, such as lithium, valproate, escitalopram,
tauopathy. Subsequently, the double mutant (K257T and tideglusib, decrease tau phosphorylation and alle-
and P301L, G272V and P301S) and triple mutant viate the process of diseases [139–142]. Currently,
(G272V, P301L, and R406W) have been reported to some of these drugs have been put into the clinical
test pathogenic hypotheses of tauopathy and thera- trials to assess the pharmacokinetics and discharge
peutic strategies [137, 138]. In the future work, more excretions. The information is provided in Table 4.
available models for human tauopathies should be No clinical benefits have been found in a phase II
produced. clinical trials for valproate in AD [143]. Escitalo-
pram reduces the levels of tau hyperphosphorylation,
TARGETING TAU THERAPEUTICS IN which are induced by A␤1-42 through the pathway
TAUOPATHIES of Akt/GSK-3␤ [144]. Other tau protein kinases are
inhibited for prevention of tau phosphorylation, but
Most tau-targeted therapies (Fig. 3) are growing no tau protein kinases inhibitors except GSK-3 have
at a rapid pace due to the discovery of the vital role been applied to clinical trials until now. To develop
of tau in neurodegeneration. Some drugs used in the other kinases inhibitors and to apply those to clini-
treatment of tauopathies have been put into clinical cal trials provide a new direction for the treatment of
phase II or III trials (Table 4), and at the same time tauopathy, but the safety and efficacy of drugs should
new drugs continue to be discovered. be taken seriously.

Fig. 3. Targeting tau-based therapeutics for tauopathies. The therapeutics of tauopathies consist of targeting phosphorylation, targeting
aggregation, increasing microtubule stabilization, tau immunization, clearance of tau, anti-inflammatory treatment, and other therapeutics.
MT, microtubule; PelA, peloruside A; NAP, daventide; NSAID, non-steroidal anti-inflammatory drugs.
C.-C. Tan et al. / Tauopathies: Mechanisms and Therapeutic Strategies 499

Table 4
Drugs of tauopathies for clinical trials
Drugs Clinical Targeting diseases ClinicalTrials.gov Conditions
phase Identifier
Lithium II PSP, CBD, AD NCT00703677 Completed
NCT01055392 Not verified
II AD NCT00088387 Completed
Valproic acid II PSP NCT00385710 Completed
0 AD NCT01729598 Completed
Tideglusib II AD NCT01350362 Discontinued
Metformin II AD NCT01965756 Ongoing
Memantine II HD, PDD, DLB NCT01458470 Completed
NCT00630500 Completed
II ALS NCT01020331 Completed
III FTD NCT00594737 Completed
IV AD NCT00800709 Completed
IV AD NCT00551161 Completed
NCT00933608* Completed
LMTX (TRx0237) III AD NCT01689233 Ongoing
III AD NCT01689246 Completed
III bvFTD NCT01626378 Completed
BMS-241027 I AD NCT01492374 Completed
NAP II PSP, CBD, FTDP-17 NCT01056965 Ongoing
AADvac1 I AD NCT01850238 Completed
I AD NCT02031198 Ongoing
II AD NCT02579252 Ongoing
C2N-8E12 I PSP NCT02494024 Ongoing
BMS-986168 I PSP NCT02460094 Ongoing
I PSP NCT02658916 Ongoing
TPI-287 I PSP, CBD NCT02133846 Ongoing
I AD NCT01966666 Ongoing
LMTX, leuo-methylthioninium; AD, Alzheimer’s disease; CBD, Corticobasal degeneration; HD,
Huntington’s disease; PDD, Parkinson’s disease dementia; PSP, Progressive supranuclear palsy;
FTD, Frontotemporal dementia; FTDP-17, Frontotemporal dementia and parkinsonism linked
to chromosome 17; NA, not available; bvFTD, Behavioral variant frontotemporal dementia.
*It includes preclinical phase.

The activation of PP2A also supplies a direction decrease the levels of tau in ex vivo slice cultures
for reducing tau phosphorylation. Sodium selenate, from transgenic mice of tauopathy [150]. The pos-
phosphotyrosyl phosphatase activator, memantine itive results have been found in a phase II trial of
and metformin reduce tau phosphorylation through methylene blue [151]. However, the National Toxi-
elevating the activity of PP2A [145–147]. Memantine cology Program used rodents to show that long-term
has been applied in clinic to attenuate the symptoms usage of methylene blue resulted in some malig-
of AD. Currently, folic acid reduces tau phosphory- nancies and lymphomas within the intestines, so
lation via inhibiting PP2A demethylation reactions leuco-methylthioninium (LMTX) is developed in
[148]. The methylation of DNA and protein plays a AD and FTD which are more bioavailable and less
key role in regulating the activity of phosphatase and toxic than MTC [152]. At present, LMTX is being
the inhibition of methylation will be of tremendous applied in several phase III studies in AD and bvFTD,
value in the treatment of tauopathies. the clinical effects will be achieved in early 2016
[151]. Recent animal studies found that anle138b
Targeting aggregation in tauopathies reduced tau aggregation, ameliorated disease symp-
toms, improved cognition, and increased survival
To inhibit or reverse tau aggregation becomes time of tau transgenic PS19 mice [153].
one of the great therapeutic strategies for neu- Natural products have also shown the function
rodegeneration. Some small molecule inhibitors are of anti-aggregation, such as oleuropein agly-
demonstrated to inhibit and disassemble tau aggrega- cone, hydroxytyrosol, oleuropein, and grape seed
tion, such as rhodanines, phenylthiazol-hydrazides, polyphenolic [154, 155]. Recently, O-GlcNAc, one
anthraquinones, benzothiazoles, phenothiazines, and modification of tau, is demonstrated to reduce the
so on [139, 149]. Methylene blue is testified to aggregation propensity of tau [156]. The above-
500 C.-C. Tan et al. / Tauopathies: Mechanisms and Therapeutic Strategies

mentioned inhibitors can prevent tau aggregation or lary pathologies in animal models [175]. Recently,
disassemble tau aggregation, but we still need much a new therapeutic strategy for passive immunization
work to do before these strategies are applied in clinic. is found to prevent phospho-tau pathology by using
encapsulated cell implants to transmit recombinant
Increasing microtubule stabilization anti-amyloid-␤ antibodies to protect against the mis-
in tauopathies folded proteins [176]. Studies on tau antibodies still
need to explore therapeutic development.
Hyperphosphorylated and polymeric forms of tau
result in a reduction of microtubule stability, and Clearance of tau in tauopathies
this reveals that keeping the stability of microtubule
is one therapeutic strategy for tauopathies. Pacli- Targeting autophagy-lysosome system
taxel, peloruside A, Epothilone D, NAP (davuentide), Autophagy-lysosomal system degrades the levels
BMS-241027 and TPI-287 improve axonal function, of abnormal tau. Therefore, enhancing the activity
MT density, and cognitive performance [157–159]. of autophagy and inhibiting the degradation of lyso-
Paclitaxel is not appropriate for human tauopathies some will be efficient to promote the degradation of
treatment because of the poor blood–brain bar- tau and alleviate neurodegeneration. Rapamycin and
rier permeability, while Epothilone D permeates the temsirolimus elevate the activity of autophagy and
blood–brain barrier. The compound makes some reduce abnormal tau in the mouse model of tauopa-
improvements in a phase II clinical trial and has thy via targeting the mTOR pathway [177, 178].
no serious effects on patients with mild to moderate Besides the mTOR pathway, other drugs also induce
AD [160]. autophagy via targeting the process of autophagy-
lysosome formation, such as trehalose, methylene
Immunization strategies for tauopathies blue, lithium, sodium valproic, carbamazepine, and
sulforaphane [150, 179–183]. The development of
Active immunization autophagy-lysosome activators that can elevate clear-
Active immunization of tau pathology decreases ance of pathological tau without negative effects will
the levels of aggregated tau through the func- be of immense therapeutic value for tauopathies and
tions of tau antigen. The phosphoepitopes of tau other proteinopathies.
are demonstrated to reduce the levels of accumu-
lated tau and prevent the process of tau related Targeting ubiquitin-proteasome system
pathology via inducing the active immunization, It becomes difficult to clear aggregated proteins
such as a PHF1 phosphoepitope in P301L trans- after the dysfunction of UPS. The enhancement of
genic mouse, phosphorylated epitope S422(PS422), ubiquitin-proteasome activity promotes the process
pS396/pS404, pS231, pS212/pS214, pS202/pT205, of tau degradation. Rolipram promotes proteasome
and AADvac1 [161–164]. A phase I trial is under- activity. IU1, a deubiquitinating enzyme related to
way for pS396/pS404 by AC Immune and Janssen proteasome, puts proteasomal degradation off [184,
and other information has been reported about this 185]. CHIP, carboxy-terminus of heat shock protein
trial [165]. Tau antigen not only induces active immu- 70-interacting protein, interacts with phosphorylated
nization, but carries a risk of causing autoimmunity tau firstly. Then it transfers phosphorylated tau to
and related tauopathy. proteasome when heat shock protein 90 captures the
abnormal tau [186]. Besides, cAMP promotes degra-
Passive immunization dation of misfolded proteins by increasing the activity
Not only active immunization, but passive immu- of 26S proteasome through PKA and Rpn6 [187].
nization has an effect on the treatment of tau pathology. These two pathways interact with 19s subunit to
These antibodies consist of anti-tau antibodies, AT8, affect the activity of proteasome. Maybe, increasing
three monoclonal antibodies of tau, MC31, DA31, the levels of cAMP will be a useful way of treating
HJ9.3, HJ9.4, PHF1, PHF13, PHF6, anti-pSer413 tauopathies.
antibody, MAb86 and C2N-8E12 [166–174]. Except
these, a novel antibody-DC8E8 is found to dis- Anti-inflammatory treatments in tauopathies
tinguish mis-disordered tau with physiological tau,
and it inhibits the interaction between pathological Neuroinflammation plays a vital role in the
tau, which reduces tau oligomers and neurofibril- process of AD and other related tauopathies.
C.-C. Tan et al. / Tauopathies: Mechanisms and Therapeutic Strategies 501

Microglial activation promotes the expression of (2016YFC1305803), the National Natural Sci-
pro-inflammatory cytokines and leads to tau hyper- ence Foundation of China (81471309, 81371406,
phosphorylation and neuronal death [188]. In 81571245, and 81501103), the Shandong Provin-
tauP301L mice deficient in CX3CR1 receptor, cial Outstanding Medical Academic Professional
microglia are over-expressed and tau is hyperphos- Program, Taishan Scholars Program of Shan-
phorylated [189], thus leading to the aggregation dong Province (ts201511109, tsqn20161078, and
of tau and related tauopathies through two path- tsqn20161079), Qingdao Key Health Discipline
ways. One is related to the activity of p38 MAP Development Fund, Qingdao Outstanding Health
kinase [190], another is related to the increased Professional Development Fund, and Shandong
NRF2-ARE (nuclear factor (erythroid-derived 2)-like Provincial Collaborative Innovation Center for
2-antioxidant response element) activity to inhibit Neurodegenerative Disorders, Research Award Fund
the overactivation of microglia [189]. So, targeting for Outstanding Young and Middle-aged Scientists
CX3CL1-CX3CR1 and/or neuronal IL-1 receptor- of Shandong Province (BS2015SW006), Projects
p38 and NRF2-ARE signaling pathway is a valid of medical and health technology development
therapeutic strategy for tauopathies. Recent studies program in Shandong province (2015WSA02054).
find that SCM-198 (Leonurine) inhibits the overacti- Authors’ disclosures available online (http://
vation of microglia via NF-␬B and c-Jun N-terminal j-alz.com/manuscript-disclosures/17-0187r1).
kinase pathways [191]. Besides, immunosuppressant
FK506 reduces tau aggregation and the expression of REFERENCES
microglia. The immunophilin FKBP52 (a member of
FK506-binding protein family) directly interacts with [1] Takashima A (2013) Tauopathies and tau oligomers.
hyperphosphorylated tau to inhibit the aggregation of J Alzheimers Dis 37, 565-568.
tau [192]. [2] Frost B, Gotz J, Feany MB (2014) Connecting the dots
between tau dysfunction and neurodegeneration. Trends
Cell Biol 25, 46-53.
[3] Villemagne VL, Fodero-Tavoletti MT, Masters CL, Rowe
CONCLUSION AND OUTLOOKS CC (2015) Tau imaging: Early progress and future direc-
tions. Lancet Neurol 14, 114-124.
Tauopathies are morphologically, biochemically, [4] Spillantini MG, Goedert M (2013) Tau pathology and neu-
and clinically heterogeneous neurodegenerative dis- rodegeneration. Lancet Neurol 12, 609-622.
[5] Wang JZ, Gao X, Wang ZH (2014) The physiology and
eases defined by the accumulation of abnormal tau pathology of microtubule-associated protein tau. Essays
proteins in the brain. Although the development of Biochem 56, 111-123.
various studies, medicine, and technology has helped [6] Wang JZ, Liu F (2008) Microtubule-associated protein tau
interpret many vital aspects of the tauopathies, more in development, degeneration and protection of neurons.
Prog Neurobiol 85, 148-175.
effective treatments for tauopathies still need to be [7] Kouri N, Carlomagno Y, Baker M, Liesinger AM, Caselli
explored. Efforts to discover imaging markers with RJ, Wszolek ZK, Petrucelli L, Boeve BF, Parisi JE, Josephs
the characteristics of lower toxicity, higher kinetics, KA, Uitti RJ, Ross OA, Graff-Radford NR, DeTure MA,
and greater permeability across the blood brain bar- Dickson DW, Rademakers R (2014) Novel mutation in
MAPT exon 13 (p. N410H) causes corticobasal degener-
rier should be continued and combined with more ation. Acta Neuropathol 127, 271-282.
efficient approaches to be applied in clinical tri- [8] Ronnback A, Nennesmo I, Tuominen H, Grueninger F,
als. Given the central role of tau in tauopathies, Viitanen M, Graff C (2014) Neuropathological characteri-
zation of two siblings carrying the MAPT S305S mutation
many treatments have constantly emerged, includ-
demonstrates features resembling argyrophilic grain dis-
ing targeting phosphorylation, targeting aggregation, ease. Acta Neuropathol 127, 297-298.
increasing microtubule stabilization, tau immuniza- [9] Iovino M, Pfisterer U, Holton JL, Lashley T, Swingler
tion, clearance of tau, anti-inflammatory treatment, RJ, Calo L, Treacy R, Revesz T, Parmar M, Goedert
M, Muqit MM, Spillantini MG (2014) The novel MAPT
and other therapeutics. It still has a long way to go mutation K298E: Mechanisms of mutant tau toxicity, brain
before we obtain a drug therapy targeting multifactor pathology and tau expression in induced fibroblast-derived
mechanisms. neurons. Acta Neuropathol 127, 283-295.
[10] Iyer A, Lapointe NE, Zielke K, Berdynski M, Guzman E,
Barczak A, Chodakowska-Zebrowska M, Barcikowska M,
ACKNOWLEDGMENTS Feinstein S, Zekanowski C (2013) A novel MAPT muta-
tion, G55R, in a frontotemporal dementia patient leads to
altered Tau function. PLoS One 8, e76409.
This work was supported by grants from [11] Graff-Radford J, Whitwell JL, Dickson DW, Josephs
the National Key R&D Program of China KA (2013) Pallidonigroluysian atrophy associated with
502 C.-C. Tan et al. / Tauopathies: Mechanisms and Therapeutic Strategies

p. A152T variant in MAPT. Parkinsonism Relat Disord Tolnay M, Uchihara T (2015) PART is part of Alzheimer
19, 838-841. disease. Acta Neuropathol 129, 749-756.
[12] Ishida C, Kobayashi K, Kitamura T, Ujike H, Iwasa K, [25] Mez J, Solomon TM, Daneshvar DH, Murphy L, Kier-
Yamada M (2015) Frontotemporal dementia with parkin- nan PT, Montenigro PH, Kriegel J, Abdolmohammadi B,
sonism linked to chromosome 17 with the MAPT R406W Fry B, Babcock KJ, Adams JW, Bourlas AP, Papadopou-
mutation presenting with a broad distribution of abundant los Z, McHale L, Ardaugh BM, Martin BR, Dixon D,
senile plaques. Neuropathology 35, 75-82. Nowinski CJ, Chaisson C, Alvarez VE, Tripodis Y, Stein
[13] Maeda S, Djukic B, Taneja P, Yu GQ, Lo I, Davis A, TD, Goldstein LE, Katz DI, Kowall NW, Cantu RC, Stern
Craft R, Guo W, Wang X, Kim D, Ponnusamy R, Gill TM, RA, McKee AC (2015) Assessing clinicopathological cor-
Masliah E, Mucke L (2016) Expression of A152T human relation in chronic traumatic encephalopathy: Rationale
tau causes age-dependent neuronal dysfunction and loss and methods for the UNITE study. Alzheimers Res Ther
in transgenic mice. EMBO Rep 17, 530-551. 7, 62.
[14] Chen J, Yu JT, Wojta K, Wang HF, Zetterberg H, Blennow [26] Stein TD, Alvarez VE, McKee AC (2014) Chronic trau-
K, Yokoyama JS, Weiner MW, Kramer JH, Rosen H, matic encephalopathy: A spectrum of neuropathological
Miller BL, Coppola G, Boxer AL (2017) Genome- changes following repetitive brain trauma in athletes and
wide association study identifies MAPT locus influencing military personnel. Alzheimers Res Ther 6, 4.
human plasma tau levels. Neurology 88, 669-676. [27] McKee AC, Stern RA, Nowinski CJ, Stein TD, Alvarez
[15] Caffrey TM, Joachim C, Wade-Martins R (2008) VE, Daneshvar DH, Lee HS, Wojtowicz SM, Hall G,
Haplotype-specific expression of the N-terminal exons 2 Baugh CM, Riley DO, Kubilus CA, Cormier KA, Jacobs
and 3 at the human MAPT locus. Neurobiol Aging 29, MA, Martin BR, Abraham CR, Ikezu T, Reichard RR,
1923-1929. Wolozin BL, Budson AE, Goldstein LE, Kowall NW,
[16] Koolen DA, Kramer JM, Neveling K, Nillesen WM, Cantu RC (2013) The spectrum of disease in chronic trau-
Moore-Barton HL, Elmslie FV, Toutain A, Amiel J, Malan matic encephalopathy. Brain 136, 43-64.
V, Tsai AC, Cheung SW, Gilissen C, Verwiel ET, Martens [28] Armstrong MJ, Litvan I, Lang AE, Bak TH, Bhatia KP,
S, Feuth T, Bongers EM, de Vries P, Scheffer H, Vissers Borroni B, Boxer AL, Dickson DW, Grossman M, Hallett
LE, de Brouwer AP, Brunner HG, Veltman JA, Schenck M, Josephs KA, Kertesz A, Lee SE, Miller BL, Reich SG,
A, Yntema HG, de Vries BB (2012) Mutations in the Riley DE, Tolosa E, Troster AI, Vidailhet M, Weiner WJ
chromatin modifier gene KANSL1 cause the 17q21.31 (2013) Criteria for the diagnosis of corticobasal degener-
microdeletion syndrome. Nat Genet 44, 639-641. ation. Neurology 80, 496-503.
[17] Tacik P, Sanchez-Contreras M, Rademakers R, Dick- [29] Murray B, Lynch T, Farrell M (2005) Clinicopathological
son DW, Wszolek ZK (2016) Genetic disorders with tau features of the tauopathies. Biochem Soc Trans 33, 595-
pathology: A review of the literature and report of two 599.
patients with tauopathy and positive family histories. Neu- [30] Ikeda C, Yokota O, Nagao S, Ishizu H, Morisada Y, Terada
rodegener Dis 16, 12-21. S, Nakashima Y, Akiyama H, Uchitomi Y (2014) Cor-
[18] Kovacs GG (2015) Invited review: Neuropathology of ticobasal degeneration initially developing motor versus
tauopathies: Principles and practice. Neuropathol Appl non-motor symptoms: A comparative clinicopathological
Neurobiol 41, 3-23. study. Psychogeriatrics 14, 152-164.
[19] Irwin DJ (2016) Tauopathies as clinicopathological enti- [31] Shimohata T, Aiba I, Nishizawa M (2015) [Criteria for the
ties. Parkinsonism Relat Disord 22(Suppl 1), S29-S33. diagnosis of corticobasal degeneration]. Brain Nerve 67,
[20] Gao Y, Tan L, Yu JT, Tan L (2017) Tau in 513-523.
Alzheimer’s disease: Mechanisms and therapeutic strate- [32] Kouri N, Murray ME, Hassan A, Rademakers R, Uitti
gies. Curr Alzheimer Res, doi: 10.2174-1567205014666 RJ, Boeve BF, Graff-Radford NR, Wszolek ZK, Litvan
170417111859 I, Josephs KA, Dickson DW (2011) Neuropathological
[21] Jiang T, Zhang YD, Chen Q, Gao Q, Zhu XC, Zhou features of corticobasal degeneration presenting as cor-
JS, Shi JQ, Lu H, Tan L, Yu JT (2016) TREM2 mod- ticobasal syndrome or Richardson syndrome. Brain 134,
ifies microglial phenotype and provides neuroprotection 3264-3275.
in P301S tau transgenic mice. Neuropharmacology 105, [33] Williams DR, Lees AJ (2009) Progressive supranuclear
196-206. palsy: Clinicopathological concepts and diagnostic chal-
[22] Scheltens P, Blennow K, Breteler MMB, de Strooper lenges. Lancet Neurol 8, 270-279.
B, Frisoni GB, Salloway S, Van der Flier WM (2016) [34] Steele JC, Richardson JC, Olszewski J (1964) Progressive
Alzheimer’s disease. Lancet 388, 505-517. supranuclear palsy. a heterogeneous degeneration involv-
[23] Crary JF, Trojanowski JQ, Schneider JA, Abisambra JF, ing the brain stem, basal ganglia and cerebellum with
Abner EL, Alafuzoff I, Arnold SE, Attems J, Beach TG, vertical gaze and pseudobulbar palsy, nuchal dystonia and
Bigio EH, Cairns NJ, Dickson DW, Gearing M, Grinberg dementia. Arch Neurol 10, 333-359.
LT, Hof PR, Hyman BT, Jellinger K, Jicha GA, Kovacs [35] Dickson DW, Ahmed Z, Algom AA, Tsuboi Y, Josephs KA
GG, Knopman DS, Kofler J, Kukull WA, Mackenzie IR, (2010) Neuropathology of variants of progressive supranu-
Masliah E, McKee A, Montine TJ, Murray ME, Neltner clear palsy. Curr Opin Neurol 23, 394-400.
JH, Santa-Maria I, Seeley WW, Serrano-Pozo A, Shelanski [36] Williams DR, Lees AJ (2010) What features improve
ML, Stein T, Takao M, Thal DR, Toledo JB, Troncoso the accuracy of the clinical diagnosis of progressive
JC, Vonsattel JP, White CL, 3rd, Wisniewski T, Woltjer supranuclear palsy-parkinsonism (PSP-P)? Mov Disord
RL, Yamada M, Nelson PT (2014) Primary age-related 25, 357-362.
tauopathy (PART): A common pathology associated with [37] Braak H, Braak E (1987) Argyrophilic grains: Character-
human aging. Acta Neuropathol 128, 755-766. istic pathology of cerebral cortex in cases of adult onset
[24] Duyckaerts C, Braak H, Brion JP, Buee L, Del Tredici dementia without Alzheimer changes. Neurosci Lett 76,
K, Goedert M, Halliday G, Neumann M, Spillantini MG, 124-127.
C.-C. Tan et al. / Tauopathies: Mechanisms and Therapeutic Strategies 503

[38] Ferrer I, Santpere G, van Leeuwen FW (2008) Argy- Vossel KA, Kramer JH, Gorno-Tempini ML, Miller BL,
rophilic grain disease. Brain 131, 1416-1432. Jagust WJ, Rabinovici GD (2016) Tau PET patterns mir-
[39] Irwin DJ, Brettschneider J, McMillan CT, Cooper F, Olm ror clinical and neuroanatomical variability in Alzheimer’s
C, Arnold SE, Van Deerlin VM, Seeley WW, Miller BL, disease. Brain 139, 1551-1567.
Lee EB, Lee VM, Grossman M, Trojanowski JQ (2015) [54] James OG, Doraiswamy PM, Borges-Neto S (2015) PET
Deep clinical and neuropathological phenotyping of Pick’s imaging of tau pathology in Alzheimer’s disease and
disease. Ann Neurol 79, 272-287. tauopathies. Front Neurol 6, 38.
[40] Kovacs GG, Rozemuller AJ, van Swieten JC, Gelpi E, [55] Hoglund K, Fourier A, Perret-Liaudet A, Zetterberg H,
Majtenyi K, Al-Sarraj S, Troakes C, Bodi I, King A, Hor- Blennow K, Portelius E (2015) Alzheimer’s disease -
tobagyi T, Esiri MM, Ansorge O, Giaccone G, Ferrer I, recent biomarker developments in relation to updated diag-
Arzberger T, Bogdanovic N, Nilsson T, Leisser I, Ala- nostic criteria. Clin Chim Acta 449, 3-8.
fuzoff I, Ironside JW, Kretzschmar H, Budka H (2013) [56] Khan TK, Alkon DL (2015) Alzheimer’s disease cere-
Neuropathology of the hippocampus in FTLD-Tau with brospinal fluid and neuroimaging biomarkers: Diagnostic
Pick bodies: A study of the BrainNet Europe Consortium. accuracy and relationship to drug efficacy. J Alzheimers
Neuropathol Appl Neurobiol 39, 166-178. Dis 46, 817-836.
[41] Irwin DJ, Cairns NJ, Grossman M, McMillan CT, [57] Struyfs H, Niemantsverdriet E, Goossens J, Fransen
Lee EB, Van Deerlin VM, Lee VM, Trojanowski JQ E, Martin JJ, De Deyn PP, Engelborghs S (2015)
(2015) Frontotemporal lobar degeneration: Defining phe- Cerebrospinal fluid p-tau181P: Biomarker for improved
notypic diversity through personalized medicine. Acta differential dementia diagnosis. Front Neurol 6, 138.
Neuropathol 129, 469-491. [58] Borroni B, Benussi A, Archetti S, Galimberti D, Parnetti
[42] Takeda N, Kishimoto Y, Yokota O (2012) Pick’s disease. L, Nacmias B, Sorbi S, Scarpini E, Padovani A (2014)
Adv Exp Med Biol 724, 300-316. Csf p-tau/tau ratio as biomarker for TDP pathology in
[43] Rockenstein E, Overk CR, Ubhi K, Mante M, Patrick frontotemporal dementia. Amyotroph Lateral Scler Fron-
C, Adame A, Bisquert A, Trejo-Morales M, Spencer B, totemporal Degener 16, 1-6.
Masliah E (2015) A novel triple repeat mutant tau trans- [59] Amadoro G, Corsetti V, Sancesario GM, Lubrano A, Mel-
genic model that mimics aspects of pick’s disease and chiorri G, Bernardini S, Calissano P, Sancesario G (2014)
fronto-temporal tauopathies. PLoS One 10, e0121570. Cerebrospinal fluid levels of a 20-22 kDa NH2 fragment
[44] Tacik P, DeTure M, Hinkle KM, Lin WL, Sanchez- of human tau provide a novel neuronal injury biomarker
Contreras M, Carlomagno Y, Pedraza O, Rademakers R, in Alzheimer’s disease and other dementias. J Alzheimers
Ross OA, Wszolek ZK, Dickson DW (2015) A novel tau Dis 42, 211-226.
mutation in Exon 12, p. Q336H, causes hereditary Pick [60] Neumann K, Farias G, Slachevsky A, Perez P, Maccioni
disease. J Neuropathol Exp Neurol 74, 1042-1052. RB (2011) Human platelets tau: A potential peripheral
[45] Ariza M, Kolb HC, Moechars D, Rombouts F, Andres JI marker for Alzheimer’s disease. J Alzheimers Dis 25,
(2015) Tau positron emission tomography (PET) imaging: 103-109.
Past, present, and future. J Med Chem 58, 4365-4382. [61] Neher MD, Keene CN, Rich MC, Moore HB, Stahel PF
[46] Villemagne VL, Okamura N (2014) In vivo tau imaging: (2014) Serum biomarkers for traumatic brain injury. South
Obstacles and progress. Alzheimers Dement 10, S254- Med J 107, 248-255.
S264. [62] Inekci D, Linemann T, Ripova D, Henriksen K, Bartos A
[47] Okamura N, Furumoto S, Fodero-Tavoletti MT, Mulligan (2014) Validation of a caspase-3 generated tau fragment as
RS, Harada R, Yates P, Pejoska S, Kudo Y, Masters CL, a serum biomarker of brain damage due to stroke: Impli-
Yanai K, Rowe CC, Villemagne VL (2014) Non-invasive cations for Alzheimer’s disease. Alzheimers Dement 10,
assessment of Alzheimer’s disease neurofibrillary pathol- P654.
ogy using 18F-THK5105 PET. Brain 137, 1762-1771. [63] Shahim P, Linemann T, Inekci D, Karsdal MA, Blennow
[48] Watanabe H, Ono M, Saji H (2015) Novel PET/SPECT K, Tegner Y, Zetterberg H, Henriksen K (2015) Serum tau
probes for imaging of tau in Alzheimer’s disease. Scien- fragments predict return to play in concussed professional
tificWorldJournal 2015, 124192. ice hockey players. J Neurotrauma 33, 1995-1999.
[49] Okamura N, Harada R, Furumoto S, Arai H, Yanai K, [64] Mattsson N, Zetterberg H, Janelidze S, Insel PS, Andreas-
Kudo Y (2014) Tau PET imaging in Alzheimer’s disease. son U, Stomrud E, Palmqvist S, Baker D, Tan Hehir
Curr Neurol Neurosci Rep 14, 500. CA, Jeromin A, Hanlon D, Song L, Shaw LM, Tro-
[50] Chien DT, Szardenings AK, Bahri S, Walsh JC, Mu F, janowski JQ, Weiner MW, Hansson O, Blennow K
Xia C, Shankle WR, Lerner AJ, Su MY, Elizarov A, Kolb (2016) Plasma tau in Alzheimer disease. Neurology 87,
HC (2014) Early clinical PET imaging results with the 1827-1835.
novel PHF-tau radioligand [F18]-T808. J Alzheimers Dis [65] Neselius S, Zetterberg H, Blennow K, Randall J, Wilson
38, 171-184. D, Marcusson J, Brisby H (2013) Olympic boxing is asso-
[51] Shao X, Carpenter GM, Desmond TJ, Sherman P, Quesada ciated with elevated levels of the neuronal protein tau in
CA, Fawaz M, Brooks AF, Kilbourn MR, Albin RL, Frey plasma. Brain Inj 27, 425-433.
KA, Scott PJ (2012) Evaluation of [(11)C]N-methyl lan- [66] Galimberti D, Villa C, Fenoglio C, Serpente M, Ghezzi L,
soprazole as a radiopharmaceutical for PET imaging of tau Cioffi SM, Arighi A, Fumagalli G, Scarpini E (2014) Cir-
neurofibrillary tangles. ACS Med Chem Lett 3, 936-941. culating miRNAs as potential biomarkers in Alzheimer’s
[52] Dani M, Brooks DJ, Edison P (2016) Tau imaging in neu- disease. J Alzheimers Dis 42, 1261-1267.
rodegenerative diseases. Eur J Nucl Med Mol Imaging 43, [67] Crespo-Biel N, Theunis C, Borghgraef P, Lechat B, Devi-
1139-1150. jver H, Maurin H, Van Leuven F (2014) Phosphorylation of
[53] Ossenkoppele R, Schonhaut DR, Scholl M, Lockhart SN, protein Tau by GSK3␤ prolongs survival of bigenic Tau.
Ayakta N, Baker SL, O’Neil JP, Janabi M, Lazaris A, P301L×GSK3␤ mice by delaying brainstem tauopathy.
Cantwell A, Vogel J, Santos M, Miller ZA, Bettcher BM, Neurobiol Dis 67, 119-132.
504 C.-C. Tan et al. / Tauopathies: Mechanisms and Therapeutic Strategies

[68] Li L, Wang W, Welford S, Zhang T, Wang X, Zhu X (2014) MicroRNA-125b induces tau hyperphosphorylation and
Ionizing radiation causes increased tau phosphorylation in cognitive deficits in Alzheimer’s disease. EMBO J 33,
primary neurons. J Neurochem 131, 86-93. 1667-1680.
[69] Mondragon-Rodriguez S, Perry G, Luna-Munoz J, [83] Salta E, Sierksma A, Vanden Eynden E, De Strooper B
Acevedo-Aquino MC, Williams S (2014) Phosphorylation (2016) miR-132 loss de-represses ITPKB and aggravates
of tau protein at sites Ser(396-404) is one of the earli- amyloid and TAU pathology in Alzheimer’s brain. EMBO
est events in Alzheimer’s disease and Down syndrome. Mol Med 8, 1005-1018.
Neuropathol Appl Neurobiol 40, 121-135. [84] Chanu SI, Sarkar S (2016) Targeted downregulation
[70] Bailey RM, Covy JP, Melrose HL, Rousseau L, Watkinson of dMyc suppresses pathogenesis of human neuronal
R, Knight J, Miles S, Farrer MJ, Dickson DW, Gias- tauopathies in drosophila by limiting heterochromatin
son BI, Lewis J (2013) LRRK2 phosphorylates novel tau relaxation and tau hyperphosphorylation. Mol Neurobiol
epitopes and promotes tauopathy. Acta Neuropathol 126, 54, 2706-2719.
809-827. [85] Inoue M, Kaida S, Nakano S, Annoni C, Nakata E, Konno
[71] Arif M, Kazim SF, Grundke-Iqbal I, Garruto RM, Iqbal T, Morii T (2014) Phosphorylation regulates fibrillation
K (2014) Tau pathology involves protein phosphatase 2A of an aggregation core peptide in the second repeat of
in parkinsonism-dementia of Guam. Proc Natl Acad Sci microtubule-binding domain of human tau. Bioorg Med
U S A 111, 1144-1149. Chem 22, 6471-6480.
[72] Dong DW, Zhang YS, Yang WY, Wang-Qin RQ, Xu AD, [86] Lim S, Haque MM, Kim D, Kim DJ, Kim YK (2014)
Ruan YW (2014) Hyperphosphorylation of tau protein in Cell-based models to investigate tau aggregation. Comput
the ipsilateral thalamus after focal cortical infarction in Struct Biotechnol J 12, 7-13.
rats. Brain Res 1543, 280-289. [87] Elbaum-Garfinkle S, Ramlall T, Rhoades E (2010) The
[73] Giraldo E, Lloret A, Fuchsberger T, Vina J (2014) Abeta role of the lipid bilayer in tau aggregation. Biophys J 98,
and tau toxicities in Alzheimer’s are linked via oxidative 2722-2730.
stress-induced p38 activation: Protective role of vitamin [88] Min SW, Cho SH, Zhou Y, Schroeder S, Haroutunian V,
E. Redox Biol 2, 873-877. Seeley WW, Huang EJ, Shen Y, Masliah E, Mukherjee C,
[74] Yang M, Lu J, Miao J, Rizak J, Yang J, Zhai R, Zhou J, Qu Meyers D, Cole PA, Ott M, Gan L (2010) Acetylation of
J, Wang J, Yang S, Ma Y, Hu X, He R (2014) Alzheimer’s tau inhibits its degradation and contributes to tauopathy.
disease and methanol toxicity (part 1): Chronic methanol Neuron 67, 953-966.
feeding led to memory impairments and tau hyperphos- [89] Song L, Lu SX, Ouyang X, Melchor J, Lee J, Terracina
phorylation in mice. J Alzheimers Dis 41, 1117-1129. G, Wang X, Hyde L, Hess JF, Parker EM, Zhang L
[75] Bodea LG, Evans HT, Van der Jeugd A, Ittner LM, Delerue (2015) Analysis of tau post-translational modifications in
F, Kril J, Halliday G, Hodges J, Kiernan MC, Gotz J (2017) rTg4510 mice, a model of tau pathology. Mol Neurode-
Accelerated aging exacerbates a pre-existing pathology in gener 10, 14.
a tau transgenic mouse model. Aging Cell 16, 377-386. [90] Cook C, Stankowski JN, Carlomagno Y, Stetler C, Petru-
[76] Yang WJ, Chen W, Chen L, Guo YJ, Zeng JS, Li GY, celli L (2014) Acetylation: A new key to unlock tau’s role
Tong WS (2017) Involvement of tau phosphorylation in in neurodegeneration. Alzheimers Res Ther 6, 29.
traumatic brain injury patients. Acta Neurol Scand 135, [91] Cook C, Gendron TF, Scheffel K, Carlomagno Y, Dun-
622-627. more J, DeTure M, Petrucelli L (2012) Loss of HDAC6, a
[77] Bretteville A, Marcouiller F, Julien C, El Khoury NB, Petry novel CHIP substrate, alleviates abnormal tau accumula-
FR, Poitras I, Mouginot D, Levesque G, Hebert SS, Planel tion. Hum Mol Genet 21, 2936-2945.
E (2012) Hypothermia-induced hyperphosphorylation: A [92] Min SW, Chen X, Tracy TE, Li Y, Zhou Y, Wang C, Shi-
new model to study tau kinase inhibitors. Sci Rep 2, 480. rakawa K, Minami SS, Defensor E, Mok SA, Sohn PD,
[78] Song B, Ao Q, Wang Z, Liu W, Niu Y, Shen Q, Zuo H, Schilling B, Cong X, Ellerby L, Gibson BW, Johnson J,
Zhang X, Gong Y (2013) Phosphorylation of tau protein Krogan N, Shamloo M, Gestwicki J, Masliah E, Verdin E,
over time in rats subjected to transient brain ischemia. Gan L (2015) Critical role of acetylation in tau-mediated
Neural Regen Res 8, 3173-3182. neurodegeneration and cognitive deficits. Nat Med 21,
[79] Fujii H, Takahashi T, Mukai T, Tanaka S, Hosomi N, 1154-1162.
Maruyama H, Sakai N, Matsumoto M (2017) Modifica- [93] Vanderweyde T, Yu H, Varnum M, Liu-Yesucevitz L, Citro
tions of tau protein after cerebral ischemia and reperfusion A, Ikezu T, Duff K, Wolozin B (2012) Contrasting pathol-
in rats are similar to those occurring in Alzheimer’s disease ogy of the stress granule proteins TIA-1 and G3BP in
- hyperphosphorylation and cleavage of 4- and 3-repeat tauopathies. J Neurosci 32, 8270-8283.
tau. J Cereb Blood Flow Metab 37, 2441-2457. [94] Ash PE, Vanderweyde TE, Youmans KL, Apicco
[80] Tiwari SS, Mizuno K, Ghosh A, Aziz W, Troakes C, Daoud DJ, Wolozin B (2014) Pathological stress granules in
J, Golash V, Noble W, Hortobagyi T, Giese KP (2016) Alzheimer’s disease. Brain Res 1584, 52-58.
Alzheimer-related decrease in CYFIP2 links amyloid pro- [95] Zilka N, Filipcik P, Koson P, Fialova L, Skrabana R,
duction to tau hyperphosphorylation and memory loss. Zilkova M, Rolkova G, Kontsekova E, Novak M (2006)
Brain 139, 2751-2765. Truncated tau from sporadic Alzheimer’s disease suffices
[81] Zhao ZB, Wu L, Xiong R, Wang LL, Zhang B, Wang to drive neurofibrillary degeneration in vivo. FEBS Lett
C, Li H, Liang L, Chen SD (2014) MicroRNA-922 pro- 580, 3582-3588.
motes tau phosphorylation by downregulating ubiquitin [96] Basurto-Islas G, Luna-Munoz J, Guillozet-Bongaarts AL,
carboxy-terminal hydrolase L1 (UCHL1) expression in the Binder LI, Mena R, Garcia-Sierra F (2008) Accumula-
pathogenesis of Alzheimer’s disease. Neuroscience 275, tion of aspartic acid421- and glutamic acid391-cleaved
232-237. tau in neurofibrillary tangles correlates with progression
[82] Banzhaf-Strathmann J, Benito E, May S, Arzberger T, in Alzheimer disease. J Neuropathol Exp Neurol 67,
Tahirovic S, Kretzschmar H, Fischer A, Edbauer D (2014) 470-483.
C.-C. Tan et al. / Tauopathies: Mechanisms and Therapeutic Strategies 505

[97] Flores-Rodriguez P, Ontiveros-Torres MA, Cardenas- ligase and ubiquitinates tau protein in vitro impair-
Aguayo MC, Luna-Arias JP, Meraz-Rios MA, ing microtubule binding. Biochim Biophys Acta 1842,
Viramontes-Pintos A, Harrington CR, Wischik CM, 1527-1538.
Mena R, Floran-Garduno B, Luna-Munoz J (2015) The [114] Shim Sang M, Lee Won J, Kim Y, Chang Jong W, Song
relationship between truncation and phosphorylation S, Jung YK (2012) Role of S5b/PSMD5 in proteasome
at the C-terminus of tau protein in the paired helical inhibition caused by TNF-␣/NFκB in higher eukaryotes.
filaments of Alzheimer’s disease. Front Neurosci 9, 33. Cell Rep 2, 603-615.
[98] Lasagna-Reeves CA, Castillo-Carranza DL, Sengupta U, [115] Suh J, Im DS, Moon GJ, Ryu KS, de Silva R, Choi IS, Lees
Clos AL, Jackson GR, Kayed R (2011) Tau oligomers AJ, Guenette SY, Tanzi RE, Gwag BJ (2010) Hypoxic
impair memory and induce synaptic and mitochondrial ischemia and proteasome dysfunction alter tau isoform
dysfunction in wild-type mice. Mol Neurodegener 6, 39. ratio by inhibiting exon 10 splicing. J Neurochem 114,
[99] Kim Y, Choi H, Lee W, Park H, Kam TI, Hong SH, Nah 160-170.
J, Jung S, Shin B, Lee H, Choi TY, Choo H, Kim KK, [116] van Tijn P, de Vrij FM, Schuurman KG, Dantuma
Choi SY, Kayed R, Jung YK (2015) Caspase-cleaved tau NP, Fischer DF, van Leeuwen FW, Hol EM (2007)
exhibits rapid memory impairment associated with tau Dose-dependent inhibition of proteasome activity by a
oligomers in a transgenic mouse model. Neurobiol Dis mutant ubiquitin associated with neurodegenerative dis-
87, 19-28. ease. J Cell Sci 120, 1615-1623.
[100] Hu Y, Li XC, Wang ZH, Luo Y, Zhang X, Liu XP, Feng Q, [117] Braak H, Thal DR, Ghebremedhin E, Del Tredici K (2011)
Wang Q, Yue Z, Chen Z, Ye K, Wang JZ, Liu GP (2016) Stages of the pathologic process in Alzheimer disease: Age
Tau accumulation impairs mitophagy via increasing mito- categories from 1 to 100 years. J Neuropathol Exp Neurol
chondrial membrane potential and reducing mitochondrial 70, 960-969.
parkin. Oncotarget 7, 17356-17368. [118] Chai X, Dage JL, Citron M (2012) Constitutive secretion
[101] Santa-Maria I, Alaniz ME, Renwick N, Cela C, Fulga TA, of tau protein by an unconventional mechanism. Neurobiol
Van Vactor D, Tuschl T, Clark LN, Shelanski ML, McCabe Dis 48, 356-366.
BD, Crary JF (2015) Dysregulation of microRNA-219 [119] Karch CM, Jeng AT, Goate AM (2012) Extracellular Tau
promotes neurodegeneration through post-transcriptional levels are influenced by variability in Tau that is associated
regulation of tau. J Clin Invest 125, 681-686. with tauopathies. J Biol Chem 287, 42751-42762.
[102] Torres-Cruz FM, Rodriguez-Cruz F, Escobar-Herrera [120] Pooler AM, Phillips EC, Lau DH, Noble W, Hanger DP
J, Barragan-Andrade N, Basurto-Islas G, Ripova D, (2013) Physiological release of endogenous tau is stimu-
Avila J, Garcia-Sierra F (2016) Expression of tau pro- lated by neuronal activity. EMBO Rep 14, 389-394.
duces aberrant plasma membrane blebbing in glial [121] Sokolow S, Henkins KM, Bilousova T, Gonzalez B, Vin-
cells through RhoA-ROCK-dependent F-actin remodel- ters HV, Miller CA, Cornwell L, Poon WW, Gylys KH
ing. J Alzheimers Dis 52, 463-482. (2014) Pre-synaptic C-terminal truncated tau is released
[103] Cuervo AM (2004) Autophagy: Many paths to the same from cortical synapses in Alzheimer’s disease. J Neu-
end. Mol Cell Biochem 263, 55-72. rochem 133, 368-379.
[104] Tan CC, Yu JT, Tan MS, Jiang T, Zhu XC, Tan L (2014) [122] Lv ZY, Tan CC, Yu JT, Tan L (2017) Spreading of pathol-
Autophagy in aging and neurodegenerative diseases: ogy in Alzheimer’s disease. Neurotox Res 32, 707-722.
Implications for pathogenesis and therapy. Neurobiol [123] Guo JL, Lee VM (2011) Seeding of normal Tau by
Aging 35, 941-957. pathological Tau conformers drives pathogenesis of
[105] Garcı́a-Arencibia M, Hochfeld WE, Toh PP, Rubinsztein Alzheimer-like tangles. J Biol Chem 286, 15317-15331.
DC (2010) Autophagy, a guardian against neurodegener- [124] Gomez-Ramos A, Diaz-Hernandez M, Rubio A, Miras-
ation. Semin Cell Dev Biol 21, 691-698. Portugal MT, Avila J (2008) Extracellular tau promotes
[106] Lee MJ, Lee JH, Rubinsztein DC (2013) Tau degradation: intracellular calcium increase through M1 and M3 mus-
The ubiquitin–proteasome system versus the autophagy- carinic receptors in neuronal cells. Mol Cell Neurosci 37,
lysosome system. Prog Neurobiol 105, 49-59. 673-681.
[107] Mizushima N (2010) The role of the Atg1/ULK1 complex [125] Avila J, Simon D, Diaz-Hernandez M, Pintor J, Hernandez
in autophagy regulation. Curr Opin Cell Biol 22, 132-139. F (2014) Sources of extracellular tau and its signaling.
[108] Kamada Y, Yoshino K, Kondo C, Kawamata T, Oshiro N, J Alzheimers Dis 40(Suppl 1), S7-S15.
Yonezawa K, Ohsumi Y (2010) Tor directly controls the [126] Wu JW, Herman M, Liu L, Simoes S, Acker CM, Figueroa
Atg1 kinase complex to regulate autophagy. Mol Cell Biol H, Steinberg JI, Margittai M, Kayed R, Zurzolo C, Di
30, 1049-1058. Paolo G, Duff KE (2013) Small misfolded Tau species
[109] Wang C, Yu JT, Miao D, Wu ZC, Tan MS, Tan L (2014) are internalized via bulk endocytosis and anterogradely
Targeting the mTOR signaling network for Alzheimer’s and retrogradely transported in neurons. J Biol Chem 288,
disease therapy. Mol Neurobiol 49, 120-135. 1856-1870.
[110] Orr M, Oddo S (2013) The role of autophagy in [127] Takeda S, Wegmann S, Cho H, DeVos SL, Commins C,
Alzheimer’s disease. Alzheimers Dement 9, P189. Roe AD, Nicholls SB, Carlson GA, Pitstick R, Nobuhara
[111] Fan W, Long Y, Lai Y, Wang X, Chen G, Zhu B (2015) CK, Costantino I, Frosch MP, Muller DJ, Irimia D, Hyman
NPAS4 facilitates the autophagic clearance of endogenous BT (2015) Neuronal uptake and propagation of a rare
tau in rat cortical neurons. J Mol Neurosci 58, 401-410. phosphorylated high-molecular-weight tau derived from
[112] Hegde AN, Upadhya SC (2011) Role of ubiquitin– Alzheimer’s disease brain. Nat Commun 6, 8490.
proteasome-mediated proteolysis in nervous system dis- [128] Liu L, Drouet V, Wu JW, Witter MP, Small SA, Clelland
ease. Biochim Biophys Acta 1809, 128-140. C, Duff K (2012) Trans-synaptic spread of tau pathology
[113] Flach K, Ramminger E, Hilbrich I, Arsalan-Werner A, in vivo. PLoS One 7, e31302.
Albrecht F, Herrmann L, Goedert M, Arendt T, Holzer [129] Clavaguera F, Hench J, Goedert M, Tolnay M (2015)
M (2014) Axotrophin/MARCH7 acts as an E3 ubiquitin Invited review: Prion-like transmission and spread-
506 C.-C. Tan et al. / Tauopathies: Mechanisms and Therapeutic Strategies

ing of tau pathology. Neuropathol Appl Neurobiol 41, beta-amyloid-induced tau hyperphosphorylation in pri-
47-58. mary hippocampal neurons through the 5-HT1A receptor
[130] Asai H, Ikezu S, Tsunoda S, Medalla M, Luebke J, Hay- mediated Akt/GSK-3beta pathway. Oncotarget 7, 13328-
dar T, Wolozin B, Butovsky O, Kugler S, Ikezu T (2015) 13339.
Depletion of microglia and inhibition of exosome synthe- [145] van Eersel J, Ke YD, Liu X, Delerue F, Kril JJ, Gotz J,
sis halt tau propagation. Nat Neurosci 18, 1584-1593. Ittner LM (2010) Sodium selenate mitigates tau pathology,
[131] Pooler AM, Polydoro M, Maury EA, Nicholls SB, Reddy neurodegeneration, and functional deficits in Alzheimer’s
SM, Wegmann S, William C, Saqran L, Cagsal-Getkin disease models. Proc Natl Acad Sci U S A 107, 13888-
O, Pitstick R, Beier DR, Carlson GA, Spires-Jones TL, 13893.
Hyman BT (2015) Amyloid accelerates tau propagation [146] Luo Y, Nie YJ, Shi HR, Ni ZF, Wang Q, Wang JZ,
and toxicity in a model of early Alzheimer’s disease. Acta Liu G-P (2013) PTPA activates protein phosphatase-2A
Neuropathol Commun 3, 14. through reducing its phosphorylation at tyrosine-307 with
[132] Dujardin S, Colin M, Buee L (2015) Invited review: upregulation of protein tyrosine phosphatase 1B. Biochim
Animal models of tauopathies and their implications Biophys Acta 1833, 1235-1243.
for research/translation into the clinic. Neuropathol Appl [147] Yoshiyama Y, Lee VM, Trojanowski JQ (2013) Therapeu-
Neurobiol 41, 59-80. tic strategies for tau mediated neurodegeneration. J Neurol
[133] Kraemer BC, Zhang B, Leverenz JB, Thomas JH, Tro- Neurosurg Psychiatry 84, 784-795.
janowski JQ, Schellenberg GD (2003) Neurodegeneration [148] Li W, Jiang M, Xiao Y, Zhang X, Cui S, Huang G (2015)
and defective neurotransmission in a Caenorhabditis ele- Folic Acid Inhibits Tau Phosphorylation through Regu-
gans model of tauopathy. Proc Natl Acad Sci U S A 100, lation of PP2A Methylation in SH-SY5Y Cells. J Nutr
9980-9985. Health Aging 19, 123-129.
[134] Fatouros C, Pir GJ, Biernat J, Koushika SP, Mandelkow E, [149] Jackson TC, Verrier JD, Kochanek PM (2013)
Mandelkow EM, Schmidt E, Baumeister R (2012) Inhibi- Anthraquinone-2-sulfonic acid (AQ2S) is a novel
tion of tau aggregation in a novel Caenorhabditis elegans neurotherapeutic agent. Cell Death Dis 4, e451.
model of tauopathy mitigates proteotoxicity. Hum Mol [150] Congdon EE, Wu JW, Myeku N, Figueroa YH, Herman
Genet 21, 3587-3603. M, Marinec PS, Gestwicki JE, Dickey CA, Yu WH, Duff
[135] Bouleau S, Tricoire H (2015) Drosophila models of KE (2012) Methylthioninium chloride (methylene blue)
Alzheimer’s disease: Advances, limits, and perspectives. induces autophagy and attenuates tauopathy in vitro and
J Alzheimers Dis 45, 1015-1038. in vivo. Autophagy 8, 609-622.
[136] Frank S, Clavaguera F, Tolnay M (2008) Tauopathy models [151] Wischik CM, Harrington CR, Storey JM (2014) Tau-
and human neuropathology: Similarities and differences. aggregation inhibitor therapy for Alzheimer’s disease.
Acta Neuropathol 115, 39-53. Biochem Pharmacol 88, 529-539.
[137] Rosenmann H, Grigoriadis N, Eldar-Levy H, Avital A, [152] Medina M, Avila J (2014) New perspectives on the role
Rozenstein L, Touloumi O, Behar L, Ben-Hur T, Avra- of tau in Alzheimer’s disease. Implications for therapy.
ham Y, Berry E, Segal M, Ginzburg I, Abramsky O (2008) Biochem Pharmacol 88, 540-547.
A novel transgenic mouse expressing double mutant tau [153] Wagner J, Krauss S, Shi S, Ryazanov S, Steffen J, Mik-
driven by its natural promoter exhibits tauopathy charac- litz C, Leonov A, Kleinknecht A, Goricke B, Weishaupt
teristics. Exp Neurol 212, 71-84. JH, Weckbecker D, Reiner AM, Zinth W, Levin J,
[138] Belarbi K, Burnouf S, Fernandez-Gomez FJ, Ehninger D, Remy S, Kretzschmar HA, Griesinger C,
Desmercieres J, Troquier L, Brouillette J, Tsam- Giese A, Fuhrmann M (2015) Reducing tau aggregates
bou L, Grosjean ME, Caillierez R, Demeyer D, Hamdane with anle138b delays disease progression in a mouse
M, Schindowski K, Blum D, Buee L (2011) Loss of model of tauopathies. Acta Neuropathol 130, 619-631.
medial septum cholinergic neurons in THY-Tau22 mouse [154] Daccache A, Lion C, Sibille N, Gerard M, Slomianny C,
model: What links with tau pathology? Curr Alzheimer Lippens G, Cotelle P (2011) Oleuropein and derivatives
Res 8, 633-638. from olives as Tau aggregation inhibitors. Neurochem Int
[139] Himmelstein DS, Ward SM, Lancia JK, Patterson KR, 58, 700-707.
Binder LI (2012) Tau as a therapeutic target in neurode- [155] Santa-Maria I, Diaz-Ruiz C, Ksiezak-Reding H, Chen A,
generative disease. Pharmacol Ther 136, 8-22. Ho L, Wang J, Pasinetti GM (2012) GSPE interferes with
[140] Hu JP, Xie JW, Wang CY, Wang T, Wang X, Wang SL, tau aggregation in vivo: Implication for treating tauopathy.
Teng WP, Wang ZY (2011) Valproate reduces tau phos- Neurobiol Aging 33, 2072-2081.
phorylation via cyclin-dependent kinase 5 and glycogen [156] Yuzwa SA, Cheung AH, Okon M, McIntosh LP,
synthase kinase 3 signaling pathways. Brain Res Bull 85, Vocadlo DJ (2014) O-GlcNAc modification of tau directly
194-200. inhibits its aggregation without perturbing the confor-
[141] Ren QG, Wang YJ, Gong WG, Zhou QD, Xu L, Zhang mational properties of tau monomers. J Mol Biol 426,
ZJ (2015) Escitalopram ameliorates forskolin-induced tau 1736-1752.
hyperphosphorylation in HEK239/tau441 cells. J Mol [157] Zhang B, Carroll J, Trojanowski JQ, Yao Y, Iba M, Potuzak
Neurosci 56, 500-508. JS, Hogan AM, Xie SX, Ballatore C, Smith AB, 3rd,
[142] Dominguez JM, Fuertes A, Orozco L, del Monte-Millan Lee VM, Brunden KR (2012) The microtubule-stabilizing
M, Delgado E, Medina M (2012) Evidence for irreversible agent, epothilone D, reduces axonal dysfunction, neuro-
inhibition of glycogen synthase kinase-3beta by tideglusib. toxicity, cognitive deficits, and Alzheimer-like pathology
J Biol Chem 287, 893-904. in an interventional study with aged tau transgenic mice.
[143] Iqbal K, Liu F, Gong CX (2015) Tau and neurodegenera- J Neurosci 32, 3601-3611.
tive disease: The story so far. Nat Rev Neurol 12, 15-27. [158] Das V, Sim DA, Miller JH (2014) Effect of taxoid
[144] Wang YJ, Ren QG, Gong WG, Wu D, Tang X, Li XL, Wu and nontaxoid site microtubule-stabilizing agents on
FF, Bai F, Xu L, Zhang ZJ (2016) Escitalopram attenuates axonal transport of mitochondria in untransfected and
C.-C. Tan et al. / Tauopathies: Mechanisms and Therapeutic Strategies 507

ECFP-htau40-transfected rat cortical neurons in culture. Barrezueta N, Polson C, Wes P, Rangan VS, Cacace A,
J Neurosci Res 92, 1155-1166. Albright CF, Meredith J Jr, Trojanowski JQ, Lee VM,
[159] Gozes I, Iram T, Maryanovsky E, Arviv C, Rozenberg L, Brunden KR, Ahlijanian M (2015) Passive immunization
Schirer Y, Giladi E, Furman-Assaf S (2014) Novel tubu- with phospho-tau antibodies reduces tau pathology and
lin and tau neuroprotective fragments sharing structural functional deficits in two distinct mouse tauopathy models.
similarities with the drug candidate NAP (Davuentide). PLoS One 10, e0125614.
J Alzheimers Dis 40(Suppl 1), S23-S36. [174] Flight MH (2013) Neurodegenerative disease: Tau
[160] Gozes I, Schirer Y, Idan-Feldman A, David M, immunotherapy targets transcellular propagation. Nat Rev
Furman-Assaf S (2014) NAP alpha-aminoisobutyric acid Drug Discov 12, 904.
(IsoNAP). J Mol Neurosci 52, 1-9. [175] Kontsekova E, Zilka N, Kovacech B, Skrabana R, Novak
[161] Wisniewski T, Goni F (2014) Immunotherapy for M (2014) Identification of structural determinants on
Alzheimer’s disease. Biochem Pharmacol 88, 499-507. tau protein essential for its pathological function: Novel
[162] Troquier L, Caillierez R, Burnouf S, Fernandez-Gomez FJ, therapeutic target for tau immunotherapy in Alzheimer’s
Grosjean ME, Zommer N, Sergeant N, Schraen-Maschke disease. Alzheimers Res Ther 6, 45.
S, Blum D, Buee L (2012) Targeting phospho-Ser422 [176] Lathuiliere A, Laversenne V, Astolfo A, Kopetzki E,
by active Tau immunotherapy in the THYTau22 mouse Jacobsen H, Stampanoni M, Bohrmann B, Schneider BL,
model: A suitable therapeutic approach. Curr Alzheimer Aebischer P (2016) A subcutaneous cellular implant for
Res 9, 397-405. passive immunization against amyloid-beta reduces brain
[163] Gruninger F (2015) Invited review: Drug development for amyloid and tau pathologies. Brain 139(Pt 5) 1587-1604.
tauopathies. Neuropathol Appl Neurobiol 41, 81-96. [177] Vidal RL, Matus S, Bargsted L, Hetz C (2014) Targeting
[164] Kontsekova E, Zilka N, Kovacech B, Novak P, Novak autophagy in neurodegenerative diseases. Trends Pharma-
M (2014) First-in-man tau vaccine targeting structural col Sci 35, 583-591.
determinants essential for pathological tau-tau interaction [178] Jiang T, Yu JT, Zhu XC, Zhang QQ, Cao L, Wang HF,
reduces tau oligomerisation and neurofibrillary degenera- Tan MS, Gao Q, Qin H, Zhang YD, Tan L (2014) Tem-
tion in an Alzheimer’s disease model. Alzheimers Res Ther sirolimus attenuates tauopathy in vitro and in vivo by
6, 44. targeting tau hyperphosphorylation and autophagic clear-
[165] Theunis C, Crespo-Biel N, Gafner V, Pihlgren M, Lopez- ance. Neuropharmacology 85, 121-130.
Deber MP, Reis P, Hickman DT, Adolfsson O, Chuard [179] Schaeffer V, Lavenir I, Ozcelik S, Tolnay M, Winkler DT,
N, Ndao DM, Borghgraef P, Devijver H, Van Leuven Goedert M (2012) Stimulation of autophagy reduces neu-
F, Pfeifer A, Muhs A (2013) Efficacy and safety of a rodegeneration in a mouse model of human tauopathy.
liposome-based vaccine against protein Tau, assessed in Brain 135, 2169-2177.
tau. P301L mice that model tauopathy. PLoS One 8, [180] Shimada K (2011) Long-term oral lithium treatment
e72301. attenuates motor disturbance by promoting autophagy in
[166] Yanamandra K, Kfoury N, Jiang H, Mahan TE, Ma S, Mal- tauopathy model mice. Alzheimers Dement 7, S483.
oney SE, Wozniak DF, Diamond MI, Holtzman DM (2013) [181] Li XZ, Chen XP, Zhao K, Bai LM, Zhang H, Zhou
Anti-tau antibodies that block tau aggregate seeding in XP (2013) Therapeutic effects of valproate combined
vitro markedly decrease pathology and improve cognition with lithium carbonate on MPTP-induced parkinsonism
in vivo. Neuron 80, 402-414. in mice: Possible mediation through enhanced autophagy.
[167] Walls KC, Ager RR, Vasilevko V, Cheng D, Medeiros R, Int J Neurosci 123, 73-79.
LaFerla FM (2014) p-Tau immunotherapy reduces soluble [182] Li L, Zhang S, Zhang X, Li T, Tang Y, Liu H, Yang W, Le
and insoluble tau in aged 3xTg-AD mice. Neurosci Lett W (2013) Autophagy enhancer carbamazepine alleviates
575, 96-100. memory deficits and cerebral amyloid-beta pathology in a
[168] Ittner A, Bertz J, Suh LS, Stevens CH, Gotz J, Ittner mouse model of Alzheimer’s disease. Curr Alzheimer Res
LM (2014) Tau-targeting passive immunization modulates 10, 433-441.
aspects of pathology in tau transgenic mice. J Neurochem [183] Cho C, Kim S, Cho SJ, Choi K, Yun SM, Koh YH, Johnson
135, 135-145. G, Park SI (2014) Sulforaphane promotes the degrada-
[169] Collin L, Bohrmann B, Gopfert U, Oroszlan-Szovik tion of phosphorylated tau via the induction of autophagy.
K, Ozmen L, Gruninger F (2014) Neuronal uptake of Alzheimers Dement 10, P476-P477.
tau/pS422 antibody and reduced progression of tau pathol- [184] Myeku N, Yu WH, Duff K (2012) Interaction between the
ogy in a mouse model of Alzheimer’s disease. Brain 137, ubiquitin proteasome system, autophagy and progressive
2834-2846. tauopathy in vivo. Alzheimers Dement 8, P636-P637.
[170] d’Abramo C, Acker CM, Jimenez HT, Davies P (2013) Tau [185] Lee BH, Lee MJ, Park S, Oh DC, Elsasser S, Chen PC,
passive immunotherapy in mutant P301L mice: Antibody Gartner C, Dimova N, Hanna J, Gygi SP, Wilson SM, King
affinity versus specificity. PLoS One 8, e62402. RW, Finley D (2010) Enhancement of proteasome activ-
[171] Yanamandra K, Jiang H, Mahan TE, Maloney SE, Woz- ity by a small-molecule inhibitor of USP14. Nature 467,
niak DF, Diamond MI, Holtzman DM (2015) Anti-tau 179-184.
antibody reduces insoluble tau and decreases brain atro- [186] Petrucelli L, Dickson D, Kehoe K, Taylor J, Snyder H,
phy. Ann Clin Transl Neurol 2, 278-288. Grover A, De Lucia M, McGowan E, Lewis J, Prihar
[172] Umeda T, Eguchi H, Kunori Y, Matsumoto Y, Taniguchi G, Kim J, Dillmann WH, Browne SE, Hall A, Voellmy
T, Mori H, Tomiyama T (2015) Passive immunotherapy R, Tsuboi Y, Dawson TM, Wolozin B, Hardy J, Hutton
of tauopathy targeting pSer413-tau: A pilot study in mice. M (2004) CHIP and Hsp70 regulate tau ubiquitination,
Ann Clin Transl Neurol 2, 241-255. degradation and aggregation. Hum Mol Genet 13, 703-714.
[173] Sankaranarayanan S, Barten DM, Vana L, Devidze N, [187] Myeku N, Clelland CL, Emrani S, Kukushkin NV, Yu WH,
Yang L, Cadelina G, Hoque N, DeCarr L, Keenan S, Lin Goldberg AL, Duff KE (2016) Tau-driven 26S proteasome
A, Cao Y, Snyder B, Zhang B, Nitla M, Hirschfeld G, impairment and cognitive dysfunction can be prevented
508 C.-C. Tan et al. / Tauopathies: Mechanisms and Therapeutic Strategies

early in disease by activating cAMP-PKA signaling. Nat pathology by the microglial fractalkine receptor. Neuron
Med 22, 46-53. 68, 19-31.
[188] Maphis N, Xu G, Kokiko-Cochran ON, Jiang S, Cardona [191] Hong ZY, Shi XR, Zhu K, Wu TT, Zhu YZ (2014)
A, Ransohoff RM, Lamb BT, Bhaskar K (2015) Reactive SCM-198 inhibits microglial overactivation and attenu-
microglia drive tau pathology and contribute to the spread- ates Abeta(1-40)-induced cognitive impairments in rats
ing of pathological tau in the brain. Brain 138, 1738-1755. via JNK and NF-small ka, CyrillicB pathways. J Neuroin-
[189] Lastres-Becker I, Innamorato NG, Jaworski T, Rabano A, flammation 11, 147.
Kugler S, Van Leuven F, Cuadrado A (2014) Fractalkine [192] Chambraud B, Sardin E, Giustiniani J, Dounane O, Schu-
activates NRF2/NFE2L2 and heme oxygenase 1 to restrain macher M, Goedert M, Baulieu EE (2010) A role for
tauopathy-induced microgliosis. Brain 137, 78-91. FKBP52 in Tau protein function. Proc Natl Acad Sci U S A
[190] Bhaskar K, Konerth M, Kokiko-Cochran ON, Cardona 107, 2658-2663.
A, Ransohoff RM, Lamb BT (2010) Regulation of tau

You might also like