Pathological Changes of Tau Related to Alzheimer's Disease - ACS

Oct 11, 2018 - Alzheimer's disease (AD), the most common form of dementia, is characterized by extracellular β-amyloid plaques and intracellular ...
2 downloads 0 Views 2MB Size
Subscriber access provided by The University of Texas at El Paso (UTEP)

Review

Pathological Changes of Tau Related to Alzheimer’s Disease Dandan Chu, and Fei Liu ACS Chem. Neurosci., Just Accepted Manuscript • DOI: 10.1021/acschemneuro.8b00457 • Publication Date (Web): 11 Oct 2018 Downloaded from http://pubs.acs.org on October 15, 2018

Just Accepted “Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted online prior to technical editing, formatting for publication and author proofing. The American Chemical Society provides “Just Accepted” as a service to the research community to expedite the dissemination of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts appear in full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been fully peer reviewed, but should not be considered the official version of record. They are citable by the Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered to authors. Therefore, the “Just Accepted” Web site may not include all articles that will be published in the journal. After a manuscript is technically edited and formatted, it will be removed from the “Just Accepted” Web site and published as an ASAP article. Note that technical editing may introduce minor changes to the manuscript text and/or graphics which could affect content, and all legal disclaimers and ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors or consequences arising from the use of information contained in these “Just Accepted” manuscripts.

is published by the American Chemical Society. 1155 Sixteenth Street N.W., Washington, DC 20036 Published by American Chemical Society. Copyright © American Chemical Society. However, no copyright claim is made to original U.S. Government works, or works produced by employees of any Commonwealth realm Crown government in the course of their duties.

Page 1 of 45 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

Pathological Changes of Tau Related to Alzheimer’s Disease Dandan Chua and Fei Liub,*

aKey

Laboratory of Neuroregeneration of Jiangsu and Ministry of Education of China,

Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226001, PR China bDepartment

of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State

Institute for Basic Research in Developmental Disabilities, Staten Island, NY 10314, USA

*Corresponding author: Address: Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY 10314, USA. Tel: 718-494-5263; Fax: 718-494-1080. [email protected].

1

ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 2 of 45

Abstract Alzheimer’s disease (AD), the most common form of dementia, is characterized by extracellular -amyloid plaques (A) and intracellular neurofibrillary tangles (NFTs), which are considered as major targets for AD therapies. However, no effective therapy is available to cure or prevent the progression of AD up till now. Accumulation of NFTs, which consist of abnormally hyperphosphorylated tau, is directly correlated with the degree of dementia in AD patients. Emerging evidence indicates that the prion-like seeding and spreading of tau pathology may be the key driver of AD. In the past decades, greater understanding of tau pathway reveals new targets for the development of specific therapies. Here, we review the recent research progress in the mechanism underlying tau pathology in AD, and briefly introduce tau-based therapeutics.

Keywords tau pathology, Alzheimer’s disease, hyperphosphorylation, truncation, aggregation, propagation

2

ACS Paragon Plus Environment

Page 3 of 45 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

1. Introduction Alzheimer’s disease (AD) is a chronic neurodegenerative disease characterized by progressive memory loss combined with problems with language and orientation, changes in mood, loss of motivation, difficulty with self-care, and impairments in thinking and behavior 1. AD is believed to be a contributor of 60-70% of dementia cases 1. The primary risk for AD is aging. Most forms of AD are sporadic, usually starting at age ~ 65 and progressing slowly over 7-10 years 2. Less than 1% of all cases are early onset familial AD, which is inherited in an autosomal dominant manner and develops similar symptoms as sporadic AD prior to age 65. Mutations of the genes for APP (amyloid precursor protein), PSEN1 (presenilin 1) and PSEN2 (presenilin 2) are responsible for the presence of familial AD 3. AD symptoms are associated with progressive loss of neurons and synapses in multiple brain regions, especially in the frontal cortex and hippocampus. The two major neuropathological hallmarks of AD are the extracellular amyloid plaques (A) and the intracellular neurofibrillary tangles (NFTs), which contain paired helical filaments (PHFs) of abnormally hyperphosphorylated tau

4-5.

In the past decades, therapies targeting Aβ did not show effective

benefit in clinical trials, casting doubt on the amyloid hypothesis of AD 6-9. The number of NFTs rather than A plaques, is positively correlated with the severity of dementia in AD patients 10. NFTs pathology is initiated in the locus coeruleus and transentorhinal area, from where it progresses to the limbic system and further to the isocortex - the Braak stages

11

- whereas the

distribution of NFTs correlates with the progression of the disease 12. Recent studies showed that tau pathology can be induced in the injection sites and anatomically connected brain regions by injection of tau aggregates isolated from AD brains or produced in vitro by incubating recombinant tau with polyanionic heparin

13-17,

similar to steps in propagation of the tau

pathology seen in AD, introducing the concept of “propagation of tau pathology”. Increasing evidence suggests that prion-like propagation of tau pathology contributes to AD progression 18,

3

ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 4 of 45

providing potential novel therapeutic strategies 7. In this review, we will discuss recent developments in tau biology and advances in tau pathological changes related to AD, and briefly introduce tau-based drug discovery.

2. Structure and function of tau Gene expression of tau Tau is one of the major microtubule-associated proteins in the brain. Human tau is encoded by the microtubule-associated protein tau (MAPT) gene, located on chromosome 17q21.31. MAPT consists of 16 exons. In the brain, the alternative splicing of exons 2, 3, and 10 produces six isoforms of tau containing zero (0N), one (1N) or two (2N) N-terminal inserts and three (3R-tau) or four (4R-tau) C-terminal microtubule-binding repeats (Fig. 1)

19.

The longest isoform of tau

consists of 441 amino acids (2N4R, tau441) with molecular weight of 45.85 kDa. The expression of tau isoforms is developmentally regulated. The fetal human brain expresses only the shortest isoform of tau (0N3R, tau352), whereas adult human brain expresses all six isoforms with approximately equal levels of 3R-tau and 4R-tau

19-20.

Rodent tau shows about 90% homology

with human tau. 3R-tau is only expressed in fetal and newborn rodents, whereas 4R-tau is mainly expressed in adult rodents 20-21. Alternative splicing of pre-mRNA is regulated by the interaction of trans-acting factors with the cis-elements. There are major two groups of trans-acting factors, serine and arginine-rich (SR) proteins and heterogeneous nuclear ribonucleoproteins (hnRNPs)

22.

Tau pre-mRNA contains

many cis-elements, including a SC35-like splicing enhancer, a polypurine enhancer (PPE) and an A/C-rich enhancer (ACE) at the 5’ end, and an exonic splicing silencer (ESS) and an exonic splicing enhancer (ESE) at the 3’ end of tau exon 10, and an intronic splicing modulator (ISM) and an intronic splicing silencer (ISS) at 5’ end of intron 10. Mutations in these cis-elements could alter the expression of tau exon 10, causing several neurodegenerative diseases 23. Splicing factors bind to these elements, and suppress exon 10 inclusion [SRSF3 (SRp20)

24,

SRSF4

(SRp75) 25, SRSF7 (9G8) 26, SRSF11 (SRp54) 27, U2AF, PTB and hnRNP G 28] or promote exon 10 inclusion [hTRA2-beta1, CELF3, CELF428-29, SRSF1 (ASF/SF2)

30,

SRSF2 (SC35)

31,

4

ACS Paragon Plus Environment

Page 5 of 45 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

SRSR6 (SRp55)

32,

protein 4 (RBM4)

30].

SRSF9 (SRp30c) 34,

RNA helicase p68 (DDX5)

and Tar DNA-binding protein 43 (TDP-43)

35

33,

RNA binding motif

promote tau exon 10

inclusion. The function of these splicing factors is modulated by phosphorylation by kinases including SR protein kinase (SRPK) (TOP1)

37,

36,

cdc-like kinase (CLK/Sty)

cAMP-dependent protein kinase (PKA)

and regulated kinase 1A (DYRK1A)

31, 39,

38,

36,

DNA topoisomerase I

dual-specificity tyrosine-phosphorylated

glycogen synthase kinase-3β (GSK-3β) 40, and AKT

serine/threonine kinase (AKT) 41. Due to the limitation of conventional PCR, studies about the amount of tau isoform transcripts in AD brain have been contradictory patients were not found in many cases reported in some studies

45, 48.

42-45.

42-44, 46-47,

Alterations of 4R-tau/3R-tau ratio in AD

while increases in 4R-tau transcripts were

Some new approaches have also been employed to analyze the

levels of tau isoform transcripts in AD. For instance, a microarray analysis of RNA isolated from single-tangle-bearing neurons detected decreased 3R-tau transcripts in AD patients

49.

Another

group used the polony profiling method to quantify individual tau isoforms, and found that a 1.3-fold increase in 4R-tau and 35% reduction in 3R-tau transcripts in AD. Among the six isoforms, 0N4R particularly accounted for the increase, while 2N3R and 1N3R were significantly decreased in AD cases

50.

The changes of protein levels of tau isoforms are more

complicated in AD. 4R-tau and 3R-tau appear equally in the early-stage and some cases of end stage AD

51-52.

In several cases of late-stage AD, 3R-tau protein is predominantly expressed in

specific brain regions with many extracellular neurofibrillary tangles (NFTs), including the subiculum, CA1 and entorhinal cortex 46, 52. These results imply that dysregulation of tau exon 10 splicing could be associated with AD progression rather than initiation. Notably, along with others, we have shown that expression of tau isoforms and total tau varies in different regions in rodent and human brains, which may cause differential vulnerability/resistance of brain regions to tau pathology 44, 53-54. So

far,

107

tau

mutations

have

been

identified

in

human

(https://www.alzforum.org/mutations). Fifteen of these mutations are associated with AD, while

5

ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 6 of 45

others are correlated to Frontotemporal dementia (FTD), Parkinsonism linked to chromosome 17 (FTDP-17), corticobasal degeneration (CBD), progressive supranuclear palsy (PSP) and other tauopathies. Tau mutations alter the peptide sequence or/and trigger the imbalance between 4R-tau and 3R-tau by changing alternative splicing 55.

Protein structure of tau Tau is an intrinsically disordered protein, which consists of many serine/threonine, proline and arginine/lysine/histidine residues, making it a highly water-soluble and basic protein with little secondary structure, and easily hyperphosphorylated. The longest isoform of human tau (tau441) contains four domains, a N-terminal projection domain which projects way from the microtubule surface, a proline-rich region responsible for the interaction of proteins with SH3 domains, a microtubule-binding domain which is involved in promoting tubulin assembly and tau aggregation, and a C-terminus (Fig. 1 and Fig. 2A)

56-57.

Tau protein is post-translationally

modified in multiple ways including phosphorylation, truncation, glycosylation, glycation, ubiquitination, nitration, methylation, lipoperoxidation, sumoylation, and acetylation

58

(Table.

1). Dysregulation of tau post-translational modifications is involved in the etiology of AD and other tauopathies.

Functions of tau Tau binds at the interface between tubulin heterodimers through its microtubule-binding repeats thereby stabilizing microtubules in vitro 59. 4R-tau isoforms have a greater affinity than 3R-tau isoforms, and are more efficient at promoting microtubule assembly 60. In normal fetal neurons, tau is enriched on dynamic microtubules in the distal axon elongation of the labile domain of microtubules

63.

55, 61-62,

where it enables the

In AD brain, pathological tau would detach

from microtubules, leaving them sensitive to severing proteins like katanin, resulting in a dramatic loss of microtubule mass 64.

6

ACS Paragon Plus Environment

Page 7 of 45 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

Additionally, tau can regulate intracellular transport along the axon

65-66.

In physiological

conditions, little tau is detected in dendrites, where it is involved in modulating postsynaptic receptor activity

67-68.

Tau is also found in the nucleus, most likely to protect the integrity of

DNA in stress response

69.

Lastly, tau is expressed at low levels in oligodendrocytes and

astrocytes. However, neuronal tau pathology is predominant in AD 70.

3. Pathological changes of tau The intracellular neurofibrillary tangle composed of abnormally hyperphosphorylated tau is one of the two neuropathological hallmarks of AD. The tau pathology starts in the coeruleus/subcoeruleus complex and the transentorhinal area (Braak stages I and II), gradually progresses toward the limbic system (Braak stages III and IV) and eventually the isocortex (Braak stages V and VI) 71. The topographic expansion of tau pathology is highly correlated with the cognitive impairment characteristics in AD, and has been used to classify the disease into six consecutive Braak stages 11.

Hyperphosphorylation of tau Tau is a phosphoprotein. The longest brain tau isoform, tau441, contains 80 serine/threonine and 5 tyrosine residues that can potentially be phosphorylated. At least 40 serine/threonine and 2 tyrosine phosphorylation sites have been identified in PHF. Normal brain tau contains 2–3 mol phosphate/mole of the protein. In AD brain, however, the phosphorylation levels of tau are 2-3-fold increased

58.

Various abnormally phosphorylated sites of tau are found in AD brain,

including Thr181, Ser195, Ser198, Ser199, Ser202, Thr205, Thr212, Ser214, Thr217, Thr231, Ser235, Ser262, Ser353, Ser396, Ser400, Ser404, Ser409 and Ser422 7. Abnormal hyperphosphorylation of tau is the key step in tau pathogenicity in AD and other tauopathies

58.

According to the phosphorylation state and solubility, tau in AD brain can be

separated into three pools: non-hyperphosphorylated functional tau (AD-tau), abnormally

7

ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 8 of 45

hyperphosphorylated tau (AD P-tau), and tau aggregated into insoluble PHFs (PHF-tau)

72.

Normal tau protein has a “paper clip” structure, in which the N- and C-terminal ends fold over the

microtubule-binding

domain

to

prevent

the

protein

from

73.

self-aggregation

Hyperphosphorylation of tau changes the protein charge and conformation, exposes the microtubule-binding domain, thereby leading to tau self-oligomerization and aggregation

58.

Approximately 40% of AD P-tau forms cytosolic oligomers that sedimented at 200,000×g 72. The major function of normal tau is to promote microtubule assembly 60. Unlike normal tau, AD P-tau can sequester normal tau as well as other microtubule-associated proteins like MAP1 and MAP2, thus disrupting microtubule assembly. However, PHF-tau has no effect on microtubule assembly, suggesting that tau oligomers and not fibrillar aggregates are cytotoxic. Dephosphorylation of AD P-tau with protein phosphatases restores tau biological activity towards microtubule assembly in vitro and diminishes tau pathology in vivo, further indicating that hyperphosphorylation is crucial in tau pathology

74-76.

In addition to effects on microtubule

stability, hyperphosphorylation of tau may influence the protein sorting, degradation, truncation, and aggregation 55. Tau

is

phosphorylated

by

proline-directed

protein

kinases

such

as

GSK-3β,

cyclin-dependent-like kinase-5 (CDK5), DYRK1A, and non-proline-directed protein kinases such as calcium/calmodulin activated protein kinase II (CaMKII), protein kinase A (PKA), casein kinase 1 (CK1), and microtubule affinity-regulated kinase 110 (MARK p110) in vivo. In AD brain, the activities or expression of these kinases are significantly altered

58, 77-83.

Tau

dephosphorylation is mediated by protein phosphatases, especially protein phosphatases 2A (PP2A), which accounts for over 70% of tau phosphatase activity in the human brain 84. In AD brain, the total tau phosphatase activity is reduced to half, which also contributes to the abnormal hyperphosphorylation of tau

84.

Therefore, the imbalance between tau kinase and phosphatase

activities is suggested to be the origin of tau hyperphosphorylation 85.

8

ACS Paragon Plus Environment

Page 9 of 45 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

Glycosylation of tau Glycosylation is an enzymatic process that covalently attaches oligosaccharides to the side chain of

a

protein.

Several

kinds

of

oligosaccharide,

including

galactose,

mannose,

N-acetylglucosamine, and sialic acid are reported to modify tau in human brain 86. Normally, tau is not modified by classic glycosylation, but is O-GlcNAcylated (with monosaccharide -N-acetylglucosamine attached to the hydroxyl groups of serine or threonine residues)

87.

However, tau from AD brain is aberrantly glycosylated through N-linkage (with glycans attached to the amide group of the asparagines residues) 86, 88. Aberrant glycosylation of tau may precede its hyperphosphorylation in AD brain, and seem to facilitate tau phosphorylation.89. In contrast, O-GlcNAcylation of tau, which negatively regulates tau phosphorylation by competing for phosphorylation sites on the protein, was decreased in AD brain 87.

Acetylation of tau Tau is acetylated by the histone acetyltransferase p300 (EP300) or CREB-binding protein (CBP), and deacetylated by sirtuin 1 (SIRT1) and histone deacetylase 6 (HDAC6) at more than twenty Lys residues within the microtubule-binding repeats and the flanking region possesses intrinsic acetyltransferase activity that allows auto-acetylation

93.

90-92.

Tau also

Acetylated-tau

pathology differs depending on the modification sites. For instance, high levels of acetylated tau at Lys163, Lys174 and Lys180 are detected in mild to moderate stages AD brains, preceding hyperphosphorylation of tau and NFT formation. Acetylation at these sites could inhibit the degradation of hyperphosphorylated tau, thus contribute to phosphorylated tau accumulation. Moreover, acetylation at Lys274, Lys280, Lys281 and Lys369 are found to impair tau function, and up-regulated in AD brain 90-91, 94-96. By contrast, Lys259, Lys290, Lys321 and Lys353 within the KXGS motifs are normally acetylated to prevent tau phosphorylation and aggregation, and are hypoacetylated in AD brain 97.

9

ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 10 of 45

Ubiquitination of tau As a natively unfolded protein, normal tau degradation could be catalyzed through an ATP/ubiquitin-independent pathway

98.

Nevertheless, tau is ubiquitylated by various ubiquitin

ligases (E3 ligases) including the C-terminus of the Hsc70-interacting protein (CHIP), TNF receptor-associated factor 6 (TRAF6) and axotrophin/MARCH7

99-101.

Mass spectrometry

reveals that soluble PHF-tau isolated from AD brains is ubiquitinated at microtubule binding domain at residues Lys254, Lys311 and Lys353

102.

Some of these residues are competitively

methylated or acetylated in AD brain 103. Tau hyperphosphorylation and N-terminal cleavage are shown to facilitate ubiquitylation of tau neurofibrillary tangle formation of tau inclusions

106-107.

104-105,

which might promote the

PHFs from AD brain are mainly monoubiquitylated rather

than polyubiquitylated, making it insufficient to induce the ubiquitin-proteasome system (UPS)-mediated proteolysis of tau aggregates

103.

Thus, enhancing polyubiquitylation and

degradation of tau might be a potential target for tau pathology 108.

SUMOylation of tau Tau can be post-translationally regulated by small ubiquitin-like modifier protein (SUMO), which is transferred to the side chain of lysine residues in a manner similar to ubiquitylation. Lys340 is the major sumoylation site of tau

109.

Sumoylation reciprocally stimulates tau

hyperphosphorylation at Tyr231 and Ser262, and inhibits tau ubiquitylation and degradation. SUMO-1 colocalizes with phosphorylated tau in AD brain and in amyloid plaques of amyloid precursor protein (APP) transgenic mice

110-111.

However, the relevance between sumoylation

and tau pathology in AD brain requires further investigation.

Glycation of tau Glycation is a nonspecific reaction of sugars and proteins, in which increased concentration of reducing sugars, generally glucose, are non-enzymatically linked to the terminal amino groups

10

ACS Paragon Plus Environment

Page 11 of 45 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

on the side chain of lysine residues. Glycation is detected in PHF-tau but not soluble tau purified from AD brains

112-113.

Glycation of tau mainly occurs in the microtubule binding domain, and

reduces its binding to microtubules, thus indirectly assists tau hyperphosphorylation and aggregation

113-115.

However, glycation alone is not sufficient to induce tau aggregation in vitro

116.

Nitration of tau Tau can be nitrated on four Tyr residues, Tyr18, Tyr29, Tyr197 and Tyr394. Nitration of all the four residues is found in AD brain; whereas Tyr197 nitration is also detected in normal brain, implying a physiological function

117-118.

Nitration of Tyr residues influences tau assembly in a

site-specific manner. Selective nitration of Tyr29 and Tyr197 increases the average length of tau filaments, whereas nitration of residues Tyr18 and Tyr394 decreases that in vitro

119.

Nevertheless, the roles of tau nitration in pathological conditions have not been extensively understood.

Truncation of tau As an intrinsically disordered protein (also known as a natively unfolded protein), tau is sensitive to protease digestion. Tau can be cleaved by calpain as well as other proteases both in vitro and in vivo

120.

Truncations of tau disrupt the “paper clip” structure of the protein and increase its

propensity to form aggregates, which can further nucleate the aggregation of normal full-length tau 55, 121. In AD brain, at least three site-specific cleavages of tau (Asn368, Glu391, and Asp421) have been identified in NFTs, and are correlated with the progression of Braak stages Similar truncations are also found in different mouse models of tauopathy

55.

122-124.

Unlike the

C-terminal truncation in NFTs, N-terminal truncations are found more associated with hyperphosphorylated high molecular weight tau oligomers (HMW-tau)

125.

Evidence has shown

that tau truncation alone is sufficient to induce hyperphosphorylation and aggregates

126.

Tau

11

ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 12 of 45

truncation can also induce toxic response like apoptosis which is independent of its function on aggregates 127. Thus, tau truncation plays a vital role in tau pathology.

Tau aggregation The natively unfolded tau monomers usually have random coil structure showing little tendency to form aggregates

128.

Partially folded forms of tau have less random coils but more -sheet

structures, which are required for the interaction between tau monomers 129-130. Two hexapeptide motifs of tau, 275VQIINK280 at the beginning of microtubule-binding repeat 2 and 306VQIVYK311 at the beginning of repeat 3, show propensity to undergo the conformational switch to -sheet structure that is responsible for the formation of aggregates

131-132.

Conversion of tau monomer

from an inert to a seed-competent form could initiate tau pathological aggregation

133.

Once the

partially folded conformation of tau monomer is triggered, they may successively aggregate together to form dimmers, soluble oligomers, insoluble PHFs and eventually NTFs 129, 134 (Fig. 2). When tau aggregates into PHFs, the repeat domains that contains β-sheet structures form the core of PHFs, while the N- and C-terminal domains contribute to a ‘fuzzy coat’ surrounding the filament core

123.

Aggregation of tau into PHFs and NFTs have been found in AD and other

related neurodegenerative diseases 135. All six isoforms of tau are detected in AD PHFs 18. Truncation and hyperphosphorylation are two major post-translational modifications that influence tau aggregation. Truncation of tau exposed the microtubule-binding repeats which are more likely to form aggregates hyperphosphorylation and aggregates

136-137.

Tau truncation alone is sufficient to induce

126, 136.

However, hyperphosphorylation of full-length tau

alone is not sufficient to induce the formation of insoluble aggregation. Only in combination with truncation could hyperphosphorylation induce tau aggregates

138.

Tau phosphorylation at

several sites (such as Thr231, Ser235, and Ser262) flanking the microtubule-binding repeats inhibits microtubule assembly and promotes its assembly into PHF hyperphosphorylation

might

facilitate

tau

aggregation

in

several

7,

139.

Generally,

ways.

Firstly,

12

ACS Paragon Plus Environment

Page 13 of 45 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

hyperphosphorylated tau detaches from microtubules, and may provide more free protein for aggregation 126. Secondly, hyperphosphorylated tau is able to sequester more normal tau into the aggregates in a prion-like manner

140.

Thirdly, hyperphosphorylation may alter tau degradation

and truncation, which indirectly affect tau aggregation 141-143. Some tau mutations affect its aggregation process. Missense mutations identified in frontotemporal dementia (FTD) and FTDP-17, including G272V, P301L, V337M and R406W, are more favorable substrate for hyperphosphorylation and are prone to form aggregates 144-145.

140,

Another example is the repeat-domain deletion mutation ΔK280 found in FTD. This

single amino acid deletion strongly reduces tau affinity for microtubules and promotes tau fibrillization by strengthening β-sheet structures 132, 145. Dysregulation of tau alternative splicing may also lead to tau aggregation. 4R-tau, which contains an extra microtubule-binding repeat, is more readily to bind and promote microtubule assembly than 3R-tau

146,

and could trigger more heparin-induced polymerization than the

combination of 3R-tau and 4R-tau do in vitro

147.

Increased 4R-tau/3R-tau ratio induces tau

phosphorylation and oligomerization in human tau-expressing mice model

148,

suggesting that

appropriate balance of 4R-tau and 3R-tau is important to prevent abnormal fibrillization. In addition, some co-factors containing strong negative charges (heparin, dextran sulfate, arachidonic acid and RNA) can induce tau aggregation in vitro

149.

For example, polyanions

(heparan sulfate) that compensate for excess positive charges of tau could cause a conformational switch of tau to form -sheet structures that facilitate aggregation

150.

Certain

proteins like 14-3-3ζ and FKBP4 are shown to induce aggregation of tau in vitro, possibly through stabilizing the aggregation-prone structure

55, 151.

Undigested products of mitochondrial

turnover may also trigger aggregation by facilitating the conformational change of tau 152.

Seeding and spreading of tau pathology Transcellular propagation of tau aggregates may drive the stereotyped progression of AD in a

13

ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 14 of 45

prion-like manner that proteopathic tau seeds are transferred from a “donor cell” to a “recipient cell” where the seeds transform normal tau into aggregates (new seeds). Once formed in the coeruleus/subcoeruleus complex, tau seeds spread along the synaptically connected neurons to target large-scale human brain networks

18, 149

(Fig. 2). However, tau pathological propagation

follows a stereotypical pattern, implying differential vulnerability of brain regions to tau pathology 54. Tau seeds isolated from AD brain (AD P-tau) mice like P301L or P301S

13, 16,

76, 153,

extracted from tau transgenic

or artificially synthesized from microtubule-binding repeats of

human tau 154 can induce propagation of tau pathology in rodent brain. Spreading of tau pathology includes the following steps: 1) uptake of tau seeds, 2) templated tau aggregation, 3) secretion and 4) transcellular transfer of tau seeds 149. Uptake of tau seeds are mediated via clathrin-independent endocytosis and macropinocytosis involving heparan sulfate proteoglycans (HSPGs) or APP in vitro

155-158.

Dynamin, actin, Cdc42, and PI3K are shown

required for the internalization of tau seeds 155. Tau seeds from diverse sources display different uptake ability. Compared to species isolated from tau transgenic mice brain or assembled from recombinant tau, the soluble HMW-tau derived from AD brain is more efficiently internalized 15. The endosomal-lysosomal and autophagy pathways are involved in the clearance of tau aggregates

152.

Due to the age-related dysfunction of these processes159 or other unknown

mechanisms, internalized tau seeds escape from endosomal vesicles and are exposed to cytoplasm, where they may template the assembly of cytoplasmic tau via direct protein-protein contact

18, 154-155.

The danger receptor galectin-8 that monitors endomembrane integrity could

protect against the release of tau seeds by recruiting NDP52-dependent autophagy machinery 155. Although tau does not contain signal peptides, it is secreted in a monomeric and/or truncated non-phosphorylated form in physiological conditions

149.

Yet the mechanism and function of

neuronal tau release are not well understood. Assembled tau aggregates are released from cells through direct leakage from the presynaptic membrane vesicles

163-164

160,

exocytosis

or direct translocation across the plasma membrane

161,

exosome

165.

162,

synaptic

Tau truncation and

14

ACS Paragon Plus Environment

Page 15 of 45 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

hyperphosphorylation favor its secretion

165-166.

Increased neuronal activity is also shown to

stimulate tau release in vitro and promote tau propagation in vivo 167. The released tau aggregates are taken up by postsynaptic neurons and subsequently template new seeds formation. Recent reports show that microglia take up both soluble and insoluble tau168. Reactive microglia is sufficient to drive tau pathology in a cell-autonomous manner 169, and may promote the spreading of tau pathology through exosome-dependent secretion

170.

These results indicate

that microglia-mediated neuroinflammation could be one possible cause for the propagation of tau pathology.

4. Tau-based therapeutics To date, only five prescription drugs are approved by the US Food and Drug Administration (FDA) for the treatment of AD. Three of these drugs (donepezil, galantamine and rivastigmine) are cholinesterase inhibitors that prevent the breakdown of acetylcholine in the brain. The fourth drug, memantine, is a non-competitive N-methyl-D-aspartate (NMDA) receptor antagonist. The fifth medication is a combination of extended-release donepezil and memantine. However, the improvement of symptoms is limited, and none of these medications prevents or slows down the progression of AD. Based on the mechanisms underlying tau pathological changes in AD, new approaches aiming at inhibiting tau pathology have been developed recently (Table. 2).

Suppression of tau gene expression Since tau is a key molecule in AD pathology, and tau knockout induces no overt phenotype except for motor and cognitive deficits in aged mice, reduction of tau expression could be a potential strategy for AD prevention 171-172. Reducing endogenous tau levels has shown to protect against A-induced cognitive impairments and behavioral abnormalities in AD model mice

173.

Antisense oligonucleotides targeting of tau diminishes neuronal hyperexcitability in drug induced mice model of seizures

174.

In AD brain, several microRNAs like miR-219, miR-125b,

15

ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 16 of 45

miR-106b, and miR-132/122 have been found to regulate tau expression and phosphorylation 175-178.

However, whether inhibition of tau expression could be beneficial in AD treatment

requires more investigation.

Inhibition of tau phosphorylation Hyperphosphorylation

determines

the

spread

of

tau

pathology.

Inhibition

of

tau

hyperphosphorylation has long been considered as a therapeutic approach, and can be accomplished through modulating the balance between tau protein kinases and phosphatases. Tau is phosphorylated by kinases such as GSK-3, CDK5, PKA and CaMKII, nevertheless, inhibition of GSK-3 or CDK5 failed in Phase II clinical trials. PP2A, the main phosphatase of tau, mediates tau dephosphorylation both directly and indirectly by regulating the phosphorylation of tau kinases. Stimulating PP2A activity shows beneficial effects on cognitive deficit in AD model rats and patients. Several medications targeting PP2A activity are currently under development or evaluation in clinical trials 7. However, we recently found that GSK-3 enhanced the methylation and activity of PP2Ac and PP2A also dephosphorylates GSK-3 at Ser9 and enhanced its kinase activity

179.

Thus, GSK-3 and PP2A regulate each other and

control tau phosphorylation both directly and indirectly through each other. Targeting either of them may affect another and attenuate its role.

Inhibition of tau aggregation Tau aggregation inhibitor therapy aims at preventing the prion-like propagation of tau pathology. Derivatives of methylene blue have been shown to disaggregate tau filaments and tangles and prevent cognition impairment in tau transgenic mice

180.

However, benefits of these drugs (such

as methylene blue, Rember TM, and LMTX) are limited in human so far. For instance, LMTX, a second-generation tau aggregation inhibitor, failed to improve cognitive and functional skills in mild to moderate AD patients in Phase III trials 7. Recently, an aggregation inhibitor NPT088 has

16

ACS Paragon Plus Environment

Page 17 of 45 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

shown to lower A plaque and phosphor-tau pathology, and to improve cognitive performance in transgenic mice

181.

NPT088 is a fusion protein that combines the backbone of a human

immunoglobulin with a general amyloid interaction motif (GAIM) which recognizes and remodels multiple misfolded proteins including both A and tau. NPT088 is currently in phase I trials. Another approach to suppress tau aggregation is to disturb the steric zippers formed by the hexapeptides.

This structure-based design of non-natural amino-acid inhibitors of tau

aggregation might have therapeutic potential as well 182.

Microtubule stabilizers In AD brain, pathological tau detaches from microtubules leading to the disruption of microtubules. Stabilization of microtubule is thus considered as a potential therapeutic approach to make up tau-induced neurotoxicity. Microtubule stabilizers were initially designed to block cancer cell mitosis because they can bind to tubulin and disturb microtubule dynamics, block mitosis in the G2/M phase, and eventually induce programmed cell apoptosis 183. Whether these promising anticancer compounds show therapeutic effects in AD treatment still requires further evaluation 7.

Immunotherapy Although the mechanism remains unclear, tau immunotherapy, either active or passive, has shown to reduce tau pathology and improve cognitive performance in tau transgenic mice and rats. Tau-based active immunotherapy which induces natural immune response achieves more persistent antibody titer levels, but might raise the risk of autoimmune reaction and other disadvantages

184.

Two vaccines, AADvac-1 which against tau peptide aa 294-305 and ACI-35

which against phosphorylated tau peptide are now in Phase II and Phase I clinical trials respectively. AADvac-1, the first active tau immunotherapy candidate tried in human, induces antibodies with high affinity to truncated tau (tau151-391). AADvac-1 active immunotherapy 17

ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 18 of 45

reduces the levels of tau oligomers and AD-like tau hyperphosphorylation, diminishes the extent of neurofibrillary pathology, and improves behavioral phenotype in transgenic rat model

185.

Although induces some side effects, AADvac-1 vaccine has a favorable safety profile and induces immune response in mild to moderate AD patients

186.

ACI-35 is a liposome-based

vaccine that carries a synthetic phosphorylated peptide to mimic phosphorylated tau at residues Ser396/Ser404. ACI-35 treatment improves the clinical phenotype and reduces tau pathology in P301L tau transgenic mice 187. However, the clinical effectiveness and safety of these vaccines in AD patients require further investigation. Compared to active immunotherapy, the passive immunotherapy is a short-term immunization achieved by continuous injection of antibody, which can be designed highly specific to target epitopes regardless of the immune variation between patients. Antibodies are generated against hyperphosphorylated tau (RO6926496, RO7105705)

188,

conformations of tau (anti-tau oligomer-specific antibody) 189-190, fragments of tau (BMS-986168, C2N-8E12)

191,

and total tau (ABBV-8E12)

192.

The therapeutic values of these antibodies are

now tested in clinical trials 7. Notably, some of these antibodies like BMS-986168, C2N-8E12 and ASN120290 might be potential therapeutic agents for other tauopathies such as progressive supranuclear palsy (PSP)

193.

We recently reported that passive immunization with tau

antibody 43D (targeting tau 6-18) decreases both tau and A pathologies and improved cognition in 3xTg-AD mice, suggesting a potential therapy for AD and related tauopathies 194-196. However, passive tau-based immunotherapy faces several challenges, one of which is the blood-brain-barrier (BBB) that allows only 0.1-0.2% of circulating antibodies to the brain. Thus technologies designed to enhance antibody delivery into the brain should also receive attentions 197.

5. Conclusions and perspectives It has been over 30 years since tau was first found to be abnormally phosphorylated in PHF in

18

ACS Paragon Plus Environment

Page 19 of 45 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

1986

4-5.

Intensive research of the protein has revealed that tau undergoes many pathological

changes in AD brain, mainly including hyperphosphorylation, truncation, aggregation, seeding and spreading. Recent studies suggest that tau oligomers, rather than fibrillar aggregates, are the cytotoxic species that disrupt synaptic function, leading to neuronal death and the propagation of tau pathology

160, 198-200.

Thus, detection and clearance of toxic tau oligomers or

even the seed-competent monomers, before the formation of PHFs and NFTs at early stages of AD, could be of crucial importance in preventing tau pathology.

Acknowledgement This work was supported in part by funds from Nantong University, New York State Office for People Developmental Disabilities and the Neural Regeneration Co-innovation Center of Jiangsu Province and by grants from the National Natural Science Foundation of China (81300978) and U.S. Alzheimer’s Association (DSAD-15-363172).

19

ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Table 1. Post-translational modifications of human tau441. Modification (ref) Sites 201 Phosphorylation T17, Y18, Y29, T39, S46, T50, T52, S56, T69, T95, T102, T123, S131, T135, S137, T149, T153, T169, T181, S184, S195, S198, S199, S202, T205, S208, T212, S214, T217, T220, T231, S235, S237, S238, T245, S258, S262, T263, S285, S289, S293, S305, S341, S352, S356, T361, T373, T386, Y394, S396, T403, S404, S409, S412, S413, T414, S416, S422, S435 O-GlcNAcylation202203

Page 20 of 45

T101, T175, S210, S241, S324, S400, S433,

T123, S208, S238, S400, S409/S412/S413*

Acetylation103

K148, K150, K163, K174, K180, K224, K225, K234, K240, K254, K257, K259, K267, K274, K280, K281, K290, K294, K298, K311, K317, K321, K331, K343, K347, K353, K369, K370, K383, K385, K395

Ubiquitination103

K254, K259, K267, K281, K290, K298, K311, K317, K321, K331, K343, K347, K353, K369, K375, K385

SUMOylation109-110

K340

Glycation103

K67, K87, K132, K148, K150, K163, K174, K180, K190, K225, K234, K259, K267, K274, K280, K281, K290, K298, K311, K317, K321, K331, K340, K343, K347, K353, K369, K370, K375, K383, K385, K395

Nitration118

Y18, Y29, Y197, Y394

Methylation103

K24, K44, K67, K87, R126, K148, K150, R155, K163, K174, K180, K190, K234, K240, K254, K259, K267, K281, K290, K311, K317, K331, R349, K353, K369, K370, K375, K385, K395 * O-GlcNAcylation may occur on any of the three sites 202.

20

ACS Paragon Plus Environment

Page 21 of 45 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47

ACS Chemical Neuroscience

Table 2. Tau-based therapeutic approaches. Approach

Name of Agent

Target type

Phase

Sponsor

Suppression of tau expression

IONIS-MAPTRx

Tau antisense oligonucleotide

1/2

Ionis Pharmaceuticals, Inc.

Inhibition of tau hyperphosphorylation

Insulin (Humulin® R U-100)

Insulin signaling activator

2/3

University of Southern California

Inhibition of tau hyperphosphorylation

Lithium carbonate

GSK-3 inhibitor

4

Ariel Gildengers

Inhibition of tau hyperphosphorylation

Metformin

Insulin sensitizer

2

University of Pennsylvania

Inhibition of tau hyperphosphorylation

Nicotinamide

Histone deacetylase inhibitor

2

University of California, Irvine

Inhibition of tau hyperphosphorylation

Nilotinb

Abl tyrosine kinase inhibitor

2

Georgetown University

Inhibition of tau hyperphosphorylation

Vorinostat

Histone deacetylase inhibitor

1

German Center for Neurodegenerative Diseases

Inhibition of tau aggregation

Methylene blue

Tau aggregation inhibitor

2

Peter Fox

Inhibition of tau aggregation

NPT088

Tau/A aggregation inhibitor

1

Proclara Biosciences, Inc.

Inhibition of tau aggregation

TRx0237

Tau aggregation inhibitor

Microtubule stabilizer

BMS-241027

Microtubule stabilizer

1

Bristol-Myers Squibb

Microtubule stabilizer

TPI-287

Microtubule stabilizer

1

University of California, San Francisco

Active immunization

AADvac 1

Synthetic tau peptide derived from a.a. 294-305

1

Axon Neuroscience SE

Active immunization

ACI-35

Synthetic phosphorylated tau peptide at S396/404

1

AC Immune SA, Janssen

Passive immunization

ABBV-8E12

Anti-tau antibody

II

AbbVie

Passive immunization

BIIB076

Anti-tau antibody

1

Biogen

Passive immunization

BMS-986168

Anti-tau antibody

2

Biogen

Passive immunization

JNJ-63733657

Anti-tau antibody

1

Janssen Research & Development, LLC

Passive immunization

LY3303560

Anti-tau antibody

2

Eli Lilly and Company

Passive immunization

RO7105705

Passive immunization against pS409

2

Genentech, Inc.

2/3

TauRx Therapeutics Ltd

21

ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 22 of 45

References 1. WHO. (2017) http://www.who.int/en/news-room/fact-sheets/detail/dementia. 2. Iqbal, K., Flory, M., Khatoon, S., Soininen, H., Pirttila, T., Lehtovirta, M., Alafuzoff, I., Blennow, K., Andreasen, N., Vanmechelen, E., and Grundke-Iqbal, I. (2005) Subgroups of Alzheimer's disease based on cerebrospinal fluid molecular markers, Ann Neurol 58, 748-757. 3. Lanoiselee, H. M., Nicolas, G., Wallon, D., Rovelet-Lecrux, A., Lacour, M., Rousseau, S., Richard, A. C., Pasquier, F., Rollin-Sillaire, A., Martinaud, O., Quillard-Muraine, M., de la Sayette, V., Boutoleau-Bretonniere, C., Etcharry-Bouyx, F., Chauvire, V., Sarazin, M., le Ber, I., Epelbaum, S., Jonveaux, T., Rouaud, O., Ceccaldi, M., Felician, O., Godefroy, O., Formaglio, M., Croisile, B., Auriacombe, S., Chamard, L., Vincent, J. L., Sauvee, M., Marelli-Tosi, C., Gabelle, A., Ozsancak, C., Pariente, J., Paquet, C., Hannequin, D., and Campion, D. (2017) APP, PSEN1, and PSEN2 mutations in early-onset Alzheimer disease: A genetic screening study of familial and sporadic cases, PLoS Med 14, e1002270. 4. Grundke-Iqbal, I., Iqbal, K., Quinlan, M., Tung, Y. C., Zaidi, M. S., and Wisniewski, H. M. (1986) Microtubule-associated protein tau. A component of Alzheimer paired helical filaments, J Biol Chem 261, 6084-6089. 5. Grundke-Iqbal, I., Iqbal, K., Tung, Y. C., Quinlan, M., Wisniewski, H. M., and Binder, L. I. (1986) Abnormal phosphorylation of the microtubule-associated protein tau (tau) in Alzheimer cytoskeletal pathology, Proc Natl Acad Sci U S A 83, 4913-4917. 6. Relkin, N. R., Thomas, R. G., Rissman, R. A., Brewer, J. B., Rafii, M. S., van Dyck, C. H., Jack, C. R., Sano, M., Knopman, D. S., Raman, R., Szabo, P., Gelmont, D. M., Fritsch, S., and Aisen, P. S. (2017) A phase 3 trial of IV immunoglobulin for Alzheimer disease, Neurology 88, 1768-1775. 7. Iqbal, K., Liu, F., and Gong, C. X. (2018) Recent developments with tau-based drug discovery, Expert Opin Drug Discov 13, 399-410. 8. Egan, M. F., Kost, J., Tariot, P. N., Aisen, P. S., Cummings, J. L., Vellas, B., Sur, C., Mukai, Y., Voss, T., Furtek, C., Mahoney, E., Harper Mozley, L., Vandenberghe, R., Mo, Y., and Michelson, D. (2018) Randomized Trial of Verubecestat for Mild-to-Moderate Alzheimer's Disease, N Engl J Med 378, 1691-1703. 9. Honig, L. S., Vellas, B., Woodward, M., Boada, M., Bullock, R., Borrie, M., Hager, K., Andreasen, N., Scarpini, E., Liu-Seifert, H., Case, M., Dean, R. A., Hake, A., Sundell, K., Poole Hoffmann, V., Carlson, C., Khanna, R., Mintun, M., DeMattos, R., Selzler, K. J., and Siemers, E. (2018) Trial of Solanezumab for Mild Dementia Due to Alzheimer's Disease, N Engl J Med 378, 321-330. 10. Arriagada, P. V., Marzloff, K., and Hyman, B. T. (1992) Distribution of Alzheimer-type pathologic changes in nondemented elderly individuals matches the pattern in Alzheimer's disease, Neurology 42, 1681-1688. 11. Braak, H., and Braak, E. (1991) Neuropathological stageing of Alzheimer-related changes, 22

ACS Paragon Plus Environment

Page 23 of 45 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

Acta Neuropathol 82, 239-259. 12. Braak, H., and Braak, E. (1995) Staging of Alzheimer's disease-related neurofibrillary changes, Neurobiol Aging 16, 271-278; discussion 278-284. 13. Clavaguera, F., Bolmont, T., Crowther, R. A., Abramowski, D., Frank, S., Probst, A., Fraser, G., Stalder, A. K., Beibel, M., Staufenbiel, M., Jucker, M., Goedert, M., and Tolnay, M. (2009) Transmission and spreading of tauopathy in transgenic mouse brain, Nat Cell Biol 11, 909-913. 14. Ahmed, Z., Cooper, J., Murray, T. K., Garn, K., McNaughton, E., Clarke, H., Parhizkar, S., Ward, M. A., Cavallini, A., Jackson, S., Bose, S., Clavaguera, F., Tolnay, M., Lavenir, I., Goedert, M., Hutton, M. L., and O'Neill, M. J. (2014) A novel in vivo model of tau propagation with rapid and progressive neurofibrillary tangle pathology: the pattern of spread is determined by connectivity, not proximity, Acta Neuropathol 127, 667-683. 15. Takeda, S., Wegmann, S., Cho, H., DeVos, S. L., Commins, C., Roe, A. D., Nicholls, S. B., Carlson, G. A., Pitstick, R., Nobuhara, C. K., Costantino, I., Frosch, M. P., Muller, D. J., Irimia, D., and Hyman, B. T. (2015) Neuronal uptake and propagation of a rare phosphorylated high-molecular-weight tau derived from Alzheimer's disease brain, Nat Commun 6, 8490. 16. Clavaguera, F., Akatsu, H., Fraser, G., Crowther, R. A., Frank, S., Hench, J., Probst, A., Winkler, D. T., Reichwald, J., Staufenbiel, M., Ghetti, B., Goedert, M., and Tolnay, M. (2013) Brain homogenates from human tauopathies induce tau inclusions in mouse brain, Proc Natl Acad Sci U S A 110, 9535-9540. 17. Boluda, S., Iba, M., Zhang, B., Raible, K. M., Lee, V. M., and Trojanowski, J. Q. (2015) Differential induction and spread of tau pathology in young PS19 tau transgenic mice following intracerebral injections of pathological tau from Alzheimer's disease or corticobasal degeneration brains, Acta Neuropathol 129, 221-237. 18. Goedert, M., Eisenberg, D. S., and Crowther, R. A. (2017) Propagation of Tau Aggregates and Neurodegeneration, Annu Rev Neurosci 40, 189-210. 19. Goedert, M., Spillantini, M. G., Jakes, R., Rutherford, D., and Crowther, R. A. (1989) Multiple isoforms of human microtubule-associated protein tau: sequences and localization in neurofibrillary tangles of Alzheimer's disease, Neuron 3, 519-526. 20. Kosik, K. S., Orecchio, L. D., Bakalis, S., and Neve, R. L. (1989) Developmentally regulated expression of specific tau sequences, Neuron 2, 1389-1397. 21. Takuma, H., Arawaka, S., and Mori, H. (2003) Isoforms changes of tau protein during development in various species, Brain Res Dev Brain Res 142, 121-127. 22. Kornblihtt, A. R., Schor, I. E., Allo, M., Dujardin, G., Petrillo, E., and Munoz, M. J. (2013) Alternative splicing: a pivotal step between eukaryotic transcription and translation, Nat Rev Mol Cell Biol 14, 153-165. 23. Qian, W., and Liu, F. (2014) Regulation of alternative splicing of tau exon 10, Neurosci Bull 30, 367-377. 23

ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 24 of 45

24. Yu, Q., Guo, J., and Zhou, J. (2004) A minimal length between tau exon 10 and 11 is required for correct splicing of exon 10, J Neurochem 90, 164-172. 25. Wang, Y., Wang, J., Gao, L., Stamm, S., and Andreadis, A. (2011) An SRp75/hnRNPG complex interacting with hnRNPE2 regulates the 5' splice site of tau exon 10, whose misregulation causes frontotemporal dementia, Gene 485, 130-138. 26. Gao, L., Wang, J., Wang, Y., and Andreadis, A. (2007) SR protein 9G8 modulates splicing of tau exon 10 via its proximal downstream intron, a clustering region for frontotemporal dementia mutations, Mol Cell Neurosci 34, 48-58. 27. Wu, J. Y., Kar, A., Kuo, D., Yu, B., and Havlioglu, N. (2006) SRp54 (SFRS11), a regulator for tau exon 10 alternative splicing identified by an expression cloning strategy, Mol Cell Biol 26, 6739-6747. 28. Andreadis, A. (2005) Tau gene alternative splicing: expression patterns, regulation and modulation of function in normal brain and neurodegenerative diseases, Biochim Biophys Acta 1739, 91-103. 29. Wang, Y., Wang, J., Gao, L., Lafyatis, R., Stamm, S., and Andreadis, A. (2005) Tau exons 2 and 10, which are misregulated in neurodegenerative diseases, are partly regulated by silencers which bind a SRp30c.SRp55 complex that either recruits or antagonizes htra2beta1, J Biol Chem 280, 14230-14239. 30. Kondo, S., Yamamoto, N., Murakami, T., Okumura, M., Mayeda, A., and Imaizumi, K. (2004) Tra2 beta, SF2/ASF and SRp30c modulate the function of an exonic splicing enhancer in exon 10 of tau pre-mRNA, Genes Cells 9, 121-130. 31. Qian, W., Liang, H., Shi, J., Jin, N., Grundke-Iqbal, I., Iqbal, K., Gong, C. X., and Liu, F. (2011) Regulation of the alternative splicing of tau exon 10 by SC35 and Dyrk1A, Nucleic Acids Res 39, 6161-6171. 32. Yin, X., Jin, N., Gu, J., Shi, J., Zhou, J., Gong, C. X., Iqbal, K., Grundke-Iqbal, I., and Liu, F. (2012) Dual-specificity tyrosine phosphorylation-regulated kinase 1A (Dyrk1A) modulates serine/arginine-rich protein 55 (SRp55)-promoted Tau exon 10 inclusion, J Biol Chem 287, 30497-30506. 33. Kar, A., Fushimi, K., Zhou, X., Ray, P., Shi, C., Chen, X., Liu, Z., Chen, S., and Wu, J. Y. (2011) RNA helicase p68 (DDX5) regulates tau exon 10 splicing by modulating a stem-loop structure at the 5' splice site, Molecular & Cellular Biology 31, 1812-1821. 34. Kar, A., Havlioglu, N., Tarn, W. Y., and Wu, J. Y. (2006) RBM4 interacts with an intronic element and stimulates tau exon 10 inclusion, Journal of Biological Chemistry 281, 24479. 35. Gu, J., Chen, F., Iqbal, K., Gong, C. X., Wang, X., and Liu, F. (2017) Transactive response DNA-binding protein 43 (TDP-43) regulates alternative splicing of tau exon 10: implications for the pathogenesis of tauopathies, Journal of Biological Chemistry 292, 10600. 36. Ngo, J. C., Chakrabarti, S., Ding, J. H., Velazquez-Dones, A., Nolen, B., Aubol, B. E., Adams, J. A., Fu, X. D., and Ghosh, G. (2005) Interplay between SRPK and Clk/Sty kinases in phosphorylation of the splicing factor ASF/SF2 is regulated by a docking motif in 24

ACS Paragon Plus Environment

Page 25 of 45 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

ASF/SF2, Mol Cell 20, 77-89. 37. Rossi, F., Labourier, E., Forne, T., Divita, G., Derancourt, J., Riou, J. F., Antoine, E., Cathala, G., Brunel, C., and Tazi, J. (1996) Specific phosphorylation of SR proteins by mammalian DNA topoisomerase I, Nature 381, 80-82. 38. Shi, J., Qian, W., Yin, X., Iqbal, K., Grundke-Iqbal, I., Gu, X., Ding, F., Gong, C. X., and Liu, F. (2011) Cyclic AMP-dependent protein kinase regulates the alternative splicing of tau exon 10: a mechanism involved in tau pathology of Alzheimer disease, J Biol Chem 286, 14639-14648. 39. Yin, X., Jin, N., Shi, J., Zhang, Y., Wu, Y., Gong, C. X., Iqbal, K., and Liu, F. (2017) Dyrk1A overexpression leads to increase of 3R-tau expression and cognitive deficits in Ts65Dn Down syndrome mice, Sci Rep 7, 619. 40. Hernandez, F., Perez, M., Lucas, J. J., Mata, A. M., Bhat, R., and Avila, J. (2004) Glycogen synthase kinase-3 plays a crucial role in tau exon 10 splicing and intranuclear distribution of SC35. Implications for Alzheimer's disease, J Biol Chem 279, 3801-3806. 41. Patel, N. A., Kaneko, S., Apostolatos, H. S., Bae, S. S., Watson, J. E., Davidowitz, K., Chappell, D. S., Birnbaum, M. J., Cheng, J. Q., and Cooper, D. R. (2005) Molecular and genetic studies imply Akt-mediated signaling promotes protein kinase CbetaII alternative splicing via phosphorylation of serine/arginine-rich splicing factor SRp40, J Biol Chem 280, 14302-14309. 42. Chambers, C. B., Lee, J. M., Troncoso, J. C., Reich, S., and Muma, N. A. (1999) Overexpression of four-repeat tau mRNA isoforms in progressive supranuclear palsy but not in Alzheimer's disease, Ann Neurol 46, 325-332. 43. Connell, J. W., Rodriguez-Martin, T., Gibb, G. M., Kahn, N. M., Grierson, A. J., Hanger, D. P., Revesz, T., Lantos, P. L., Anderton, B. H., and Gallo, J. M. (2005) Quantitative analysis of tau isoform transcripts in sporadic tauopathies, Brain Res Mol Brain Res 137, 104-109. 44. Ingelsson, M., Ramasamy, K., Cantuti-Castelvetri, I., Skoglund, L., Matsui, T., Orne, J., Kowa, H., Raju, S., Vanderburg, C. R., Augustinack, J. C., de Silva, R., Lees, A. J., Lannfelt, L., Growdon, J. H., Frosch, M. P., Standaert, D. G., Irizarry, M. C., and Hyman, B. T. (2006) No alteration in tau exon 10 alternative splicing in tangle-bearing neurons of the Alzheimer's disease brain, Acta Neuropathol 112, 439-449. 45. Glatz, D. C., Rujescu, D., Tang, Y., Berendt, F. J., Hartmann, A. M., Faltraco, F., Rosenberg, C., Hulette, C., Jellinger, K., Hampel, H., Riederer, P., Moller, H. J., Andreadis, A., Henkel, K., and Stamm, S. (2006) The alternative splicing of tau exon 10 and its regulatory proteins CLK2 and TRA2-BETA1 changes in sporadic Alzheimer's disease, J Neurochem 96, 635-644. 46. Boutajangout, A., Boom, A., Leroy, K., and Brion, J. P. (2004) Expression of tau mRNA and soluble tau isoforms in affected and non-affected brain areas in Alzheimer's disease, FEBS Lett 576, 183-189. 47. Umeda, Y., Taniguchi, S., Arima, K., Piao, Y. S., Takahashi, H., Iwatsubo, T., Mann, D., and 25

ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 26 of 45

Hasegawa, M. (2004) Alterations in human tau transcripts correlate with those of neurofilament in sporadic tauopathies, Neurosci Lett 359, 151-154. 48. Yasojima, K., McGeer, E. G., and McGeer, P. L. (1999) Tangled areas of Alzheimer brain have upregulated levels of exon 10 containing tau mRNA, Brain Res 831, 301-305. 49. Ginsberg, S. D., Che, S., Counts, S. E., and Mufson, E. J. (2006) Shift in the ratio of three-repeat tau and four-repeat tau mRNAs in individual cholinergic basal forebrain neurons in mild cognitive impairment and Alzheimer's disease, J Neurochem 96, 1401-1408. 50. Conrad, C., Zhu, J., Schoenfeld, D., Fang, Z., Ingelsson, M., Stamm, S., Church, G., and Hyman, B. T. (2007) Single molecule profiling of tau gene expression in Alzheimer's disease, J Neurochem 103, 1228-1236. 51. Togo, T., Akiyama, H., Iseki, E., Uchikado, H., Kondo, H., Ikeda, K., Tsuchiya, K., de Silva, R., Lees, A., and Kosaka, K. (2004) Immunohistochemical study of tau accumulation in early stages of Alzheimer-type neurofibrillary lesions, Acta Neuropathol 107, 504-508. 52. Espinoza, M., de Silva, R., Dickson, D. W., and Davies, P. (2008) Differential incorporation of tau isoforms in Alzheimer's disease, J Alzheimers Dis 14, 1-16. 53. Trabzuni, D., Wray, S., Vandrovcova, J., Ramasamy, A., Walker, R., Smith, C., Luk, C., Gibbs, J. R., Dillman, A., Hernandez, D. G., Arepalli, S., Singleton, A. B., Cookson, M. R., Pittman, A. M., de Silva, R., Weale, M. E., Hardy, J., and Ryten, M. (2012) MAPT expression and splicing is differentially regulated by brain region: relation to genotype and implication for tauopathies, Hum Mol Genet 21, 4094-4103. 54. Hu, W., Wu, F., Zhang, Y., Gong, C. X., Iqbal, K., and Liu, F. (2017) Expression of Tau Pathology-Related Proteins in Different Brain Regions: A Molecular Basis of Tau Pathogenesis, Front Aging Neurosci 9, 311. 55. Wang, Y., and Mandelkow, E. (2016) Tau in physiology and pathology, Nat Rev Neurosci 17, 5-21. 56. Mandelkow, E. M., Biernat, J., Drewes, G., Gustke, N., Trinczek, B., and Mandelkow, E. (1995) Tau domains, phosphorylation, and interactions with microtubules, Neurobiol Aging 16, 355-362; discussion 362-353. 57. Zempel, H., and Mandelkow, E. (2014) Lost after translation: missorting of Tau protein and consequences for Alzheimer disease, Trends Neurosci 37, 721-732. 58. Iqbal, K., Liu, F., and Gong, C. X. (2016) Tau and neurodegenerative disease: the story so far, Nat Rev Neurol 12, 15-27. 59. Kadavath, H., Hofele, R. V., Biernat, J., Kumar, S., Tepper, K., Urlaub, H., Mandelkow, E., and Zweckstetter, M. (2015) Tau stabilizes microtubules by binding at the interface between tubulin heterodimers, Proc Natl Acad Sci U S A 112, 7501-7506. 60. Goedert, M., and Jakes, R. (1990) Expression of separate isoforms of human tau protein: correlation with the tau pattern in brain and effects on tubulin polymerization, EMBO J 9, 4225-4230. 61. Black, M. M., Slaughter, T., Moshiach, S., Obrocka, M., and Fischer, I. (1996) Tau is 26

ACS Paragon Plus Environment

Page 27 of 45 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

enriched on dynamic microtubules in the distal region of growing axons, J Neurosci 16, 3601-3619. 62. Kempf, M., Clement, A., Faissner, A., Lee, G., and Brandt, R. (1996) Tau binds to the distal axon early in development of polarity in a microtubule- and microfilament-dependent manner, J Neurosci 16, 5583-5592. 63. Qiang, L., Sun, X., Austin, T., Muralidharan, H., Jean, L., Liu, M., Yu, W., and Baas, P. (2018) Tau Does Not Stabilize Axonal Microtubules but Rather Enables Them to Have Long Labile Domains, Current Biology 28, 1-9. 64. Austin, T. O., Qiang, L., and Baas, P. W. (2017) Chapter 4 – Mechanisms of Neuronal Microtubule Loss in Alzheimer’s Disease, Neuroprotection in Alzheimers Disease, 59-71. 65. Kanaan, N. M., Morfini, G. A., LaPointe, N. E., Pigino, G. F., Patterson, K. R., Song, Y., Andreadis, A., Fu, Y., Brady, S. T., and Binder, L. I. (2011) Pathogenic forms of tau inhibit kinesin-dependent axonal transport through a mechanism involving activation of axonal phosphotransferases, J Neurosci 31, 9858-9868. 66. Stamer, K., Vogel, R., Thies, E., Mandelkow, E., and Mandelkow, E. M. (2002) Tau blocks traffic of organelles, neurofilaments, and APP vesicles in neurons and enhances oxidative stress, J Cell Biol 156, 1051-1063. 67. Hirokawa, N., Funakoshi, T., Sato-Harada, R., and Kanai, Y. (1996) Selective stabilization of tau in axons and microtubule-associated protein 2C in cell bodies and dendrites contributes to polarized localization of cytoskeletal proteins in mature neurons, J Cell Biol 132, 667-679. 68. Ittner, L. M., Ke, Y. D., Delerue, F., Bi, M., Gladbach, A., van Eersel, J., Wolfing, H., Chieng, B. C., Christie, M. J., Napier, I. A., Eckert, A., Staufenbiel, M., Hardeman, E., and Gotz, J. (2010) Dendritic function of tau mediates amyloid-beta toxicity in Alzheimer's disease mouse models, Cell 142, 387-397. 69. Sultan, A., Nesslany, F., Violet, M., Begard, S., Loyens, A., Talahari, S., Mansuroglu, Z., Marzin, D., Sergeant, N., Humez, S., Colin, M., Bonnefoy, E., Buee, L., and Galas, M. C. (2011) Nuclear tau, a key player in neuronal DNA protection, J Biol Chem 286, 4566-4575. 70. Kahlson, M. A., and Colodner, K. J. (2015) Glial Tau Pathology in Tauopathies: Functional Consequences, J Exp Neurosci 9, 43-50. 71. Braak, H., and Del Tredici, K. (2011) The pathological process underlying Alzheimer's disease in individuals under thirty, Acta Neuropathol 121, 171-181. 72. Kopke, E., Tung, Y. C., Shaikh, S., Alonso, A. C., Iqbal, K., and Grundke-Iqbal, I. (1993) Microtubule-associated protein tau. Abnormal phosphorylation of a non-paired helical filament pool in Alzheimer disease, J Biol Chem 268, 24374-24384. 73. Mandelkow, E., von Bergen, M., Biernat, J., and Mandelkow, E. M. (2007) Structural principles of tau and the paired helical filaments of Alzheimer's disease, Brain Pathol 17, 83-90. 74. Wang, J. Z., Grundke-Iqbal, I., and Iqbal, K. (1996) Restoration of biological activity of Alzheimer abnormally phosphorylated tau by dephosphorylation with protein 27

ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 28 of 45

phosphatase-2A, -2B and -1, Brain Res Mol Brain Res 38, 200-208. 75. Wang, J. Z., Gong, C. X., Zaidi, T., Grundke-Iqbal, I., and Iqbal, K. (1995) Dephosphorylation of Alzheimer paired helical filaments by protein phosphatase-2A and -2B, J Biol Chem 270, 4854-4860. 76. Hu, W., Zhang, X., Tung, Y. C., Xie, S., Liu, F., and Iqbal, K. (2016) Hyperphosphorylation determines both the spread and the morphology of tau pathology, Alzheimers Dement 12, 1066-1077. 77. Jin, N., Yin, X., Gu, J., Zhang, X., Shi, J., Qian, W., Ji, Y., Cao, M., Gu, X., Ding, F., Iqbal, K., Gong, C. X., and Liu, F. (2015) Truncation and Activation of Dual Specificity Tyrosine Phosphorylation-regulated Kinase 1A by Calpain I: A MOLECULAR MECHANISM LINKED TO TAU PATHOLOGY IN ALZHEIMER DISEASE, J Biol Chem 290, 15219-15237. 78. Jin, N., Yin, X., Yu, D., Cao, M., Gong, C. X., Iqbal, K., Ding, F., Gu, X., and Liu, F. (2015) Truncation and activation of GSK-3beta by calpain I: a molecular mechanism links to tau hyperphosphorylation in Alzheimer's disease, Sci Rep 5, 8187. 79. Pei, J. J., Grundke-Iqbal, I., Iqbal, K., Bogdanovic, N., Winblad, B., and Cowburn, R. F. (1998) Accumulation of cyclin-dependent kinase 5 (cdk5) in neurons with early stages of Alzheimer's disease neurofibrillary degeneration, Brain Res 797, 267-277. 80. Cavazzin, C., Bonvicini, C., Nocera, A., Racchi, M., Kasahara, J., Tardito, D., Gennarelli, M., Govoni, S., Racagni, G., and Popoli, M. (2004) Expression and phosphorylation of delta-CaM kinase II in cultured Alzheimer fibroblasts, Neurobiol Aging 25, 1187-1196. 81. Liang, Z., Liu, F., Grundke-Iqbal, I., Iqbal, K., and Gong, C. X. (2007) Down-regulation of cAMP-dependent protein kinase by over-activated calpain in Alzheimer disease brain, J Neurochem 103, 2462-2470. 82. Chen, C., Gu, J., Basurto-Islas, G., Jin, N., Wu, F., Gong, C. X., Iqbal, K., and Liu, F. (2017) Up-regulation of casein kinase 1epsilon is involved in tau pathogenesis in Alzheimer's disease, Sci Rep 7, 13478. 83. Chin, J. Y., Knowles, R. B., Schneider, A., Drewes, G., Mandelkow, E. M., and Hyman, B. T. (2000) Microtubule-affinity regulating kinase (MARK) is tightly associated with neurofibrillary tangles in Alzheimer brain: a fluorescence resonance energy transfer study, J Neuropathol Exp Neurol 59, 966-971. 84. Liu, F., Grundke-Iqbal, I., Iqbal, K., and Gong, C. X. (2005) Contributions of protein phosphatases PP1, PP2A, PP2B and PP5 to the regulation of tau phosphorylation, Eur J Neurosci 22, 1942-1950. 85. Martin, L., Latypova, X., Wilson, C. M., Magnaudeix, A., Perrin, M. L., Yardin, C., and Terro, F. (2013) Tau protein kinases: involvement in Alzheimer's disease, Ageing Research Reviews 12, 289. 86. Liu, F., Zaidi, T., Iqbal, K., Grundke-Iqbal, I., Merkle, R. K., and Gong, C. X. (2002) Role of glycosylation in hyperphosphorylation of tau in Alzheimer's disease, FEBS Lett 512, 28

ACS Paragon Plus Environment

Page 29 of 45 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

101-106. 87. Liu, F., Iqbal, K., Grundke-Iqbal, I., Hart, G. W., and Gong, C. X. (2004) O-GlcNAcylation regulates phosphorylation of tau: a mechanism involved in Alzheimer's disease, Proc Natl Acad Sci U S A 101, 10804-10809. 88. Wang, J. Z., Grundke-Iqbal, I., and Iqbal, K. (1996) Glycosylation of microtubule-associated protein tau: an abnormal posttranslational modification in Alzheimer's disease, Nat Med 2, 871-875. 89. Gong, C. X., Liu, F., Grundke-Iqbal, I., and Iqbal, K. (2005) Post-translational modifications of tau protein in Alzheimer's disease, J Neural Transm (Vienna) 112, 813-838. 90. Min, S. W., Cho, S. H., Zhou, Y., Schroeder, S., Haroutunian, V., Seeley, W. W., Huang, E. J., Shen, Y., Masliah, E., Mukherjee, C., Meyers, D., Cole, P. A., Ott, M., and Gan, L. (2010) Acetylation of tau inhibits its degradation and contributes to tauopathy, Neuron 67, 953-966. 91. Cohen, T. J., Guo, J. L., Hurtado, D. E., Kwong, L. K., Mills, I. P., Trojanowski, J. Q., and Lee, V. M. (2011) The acetylation of tau inhibits its function and promotes pathological tau aggregation, Nat Commun 2, 252. 92. Cook, C., Stankowski, J. N., Carlomagno, Y., Stetler, C., and Petrucelli, L. (2014) Acetylation: a new key to unlock tau's role in neurodegeneration, Alzheimers Res Ther 6, 29. 93. Cohen, T. J., Friedmann, D., Hwang, A. W., Marmorstein, R., and Lee, V. M. (2013) The microtubule-associated tau protein has intrinsic acetyltransferase activity, Nat Struct Mol Biol 20, 756-762. 94. Irwin, D. J., Cohen, T. J., Grossman, M., Arnold, S. E., McCarty-Wood, E., Van Deerlin, V. M., Lee, V. M., and Trojanowski, J. Q. (2013) Acetylated tau neuropathology in sporadic and hereditary tauopathies, Am J Pathol 183, 344-351. 95. Min, S. W., Chen, X., Tracy, T. E., Li, Y., Zhou, Y., Wang, C., Shirakawa, K., Minami, S. S., Defensor, E., Mok, S. A., Sohn, P. D., Schilling, B., Cong, X., Ellerby, L., Gibson, B. W., Johnson, J., Krogan, N., Shamloo, M., Gestwicki, J., Masliah, E., Verdin, E., and Gan, L. (2015) Critical role of acetylation in tau-mediated neurodegeneration and cognitive deficits, Nat Med 21, 1154-1162. 96. Tracy, T. E., Sohn, P. D., Minami, S. S., Wang, C., Min, S. W., Li, Y., Zhou, Y., Le, D., Lo, I., Ponnusamy, R., Cong, X., Schilling, B., Ellerby, L. M., Huganir, R. L., and Gan, L. (2016) Acetylated Tau Obstructs KIBRA-Mediated Signaling in Synaptic Plasticity and Promotes Tauopathy-Related Memory Loss, Neuron 90, 245-260. 97. Cook, C., Carlomagno, Y., Gendron, T. F., Dunmore, J., Scheffel, K., Stetler, C., Davis, M., Dickson, D., Jarpe, M., DeTure, M., and Petrucelli, L. (2014) Acetylation of the KXGS motifs in tau is a critical determinant in modulation of tau aggregation and clearance, Hum Mol Genet 23, 104-116. 98. Grune, T., Botzen, D., Engels, M., Voss, P., Kaiser, B., Jung, T., Grimm, S., Ermak, G., and Davies, K. J. (2010) Tau protein degradation is catalyzed by the ATP/ubiquitin-independent 20S proteasome under normal cell conditions, Arch Biochem Biophys 500, 181-188. 29

ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 30 of 45

99. Flach, K., Ramminger, E., Hilbrich, I., Arsalan-Werner, A., Albrecht, F., Herrmann, L., Goedert, M., Arendt, T., and Holzer, M. (2014) Axotrophin/MARCH7 acts as an E3 ubiquitin ligase and ubiquitinates tau protein in vitro impairing microtubule binding, Biochim Biophys Acta 1842, 1527-1538. 100. Petrucelli, L., Dickson, D., Kehoe, K., Taylor, J., Snyder, H., Grover, A., De Lucia, M., McGowan, E., Lewis, J., Prihar, G., Kim, J., Dillmann, W. H., Browne, S. E., Hall, A., Voellmy, R., Tsuboi, Y., Dawson, T. M., Wolozin, B., Hardy, J., and Hutton, M. (2004) CHIP and Hsp70 regulate tau ubiquitination, degradation and aggregation, Hum Mol Genet 13, 703-714. 101. Babu, J. R., Geetha, T., and Wooten, M. W. (2005) Sequestosome 1/p62 shuttles polyubiquitinated tau for proteasomal degradation, J Neurochem 94, 192-203. 102. Cripps, D., Thomas, S. N., Jeng, Y., Yang, F., Davies, P., and Yang, A. J. (2006) Alzheimer disease-specific conformation of hyperphosphorylated paired helical filament-Tau is polyubiquitinated through Lys-48, Lys-11, and Lys-6 ubiquitin conjugation, J Biol Chem 281, 10825-10838. 103. Kontaxi, C., Piccardo, P., and Gill, A. C. (2017) Lysine-Directed Post-translational Modifications of Tau Protein in Alzheimer's Disease and Related Tauopathies, Front Mol Biosci 4, 56. 104. Shimura, H., Schwartz, D., Gygi, S. P., and Kosik, K. S. (2004) CHIP-Hsc70 complex ubiquitinates phosphorylated tau and enhances cell survival, J Biol Chem 279, 4869-4876. 105. Bancher, C., Grundke-Iqbal, I., Iqbal, K., Fried, V. A., Smith, H. T., and Wisniewski, H. M. (1991) Abnormal phosphorylation of tau precedes ubiquitination in neurofibrillary pathology of Alzheimer disease, Brain Res 539, 11-18. 106. Tan, J. M., Wong, E. S., Kirkpatrick, D. S., Pletnikova, O., Ko, H. S., Tay, S. P., Ho, M. W., Troncoso, J., Gygi, S. P., Lee, M. K., Dawson, V. L., Dawson, T. M., and Lim, K. L. (2008) Lysine 63-linked ubiquitination promotes the formation and autophagic clearance of protein inclusions associated with neurodegenerative diseases, Hum Mol Genet 17, 431-439. 107. Baker, M., Mackenzie, I. R., Pickering-Brown, S. M., Gass, J., Rademakers, R., Lindholm, C., Snowden, J., Adamson, J., Sadovnick, A. D., Rollinson, S., Cannon, A., Dwosh, E., Neary, D., Melquist, S., Richardson, A., Dickson, D., Berger, Z., Eriksen, J., Robinson, T., Zehr, C., Dickey, C. A., Crook, R., McGowan, E., Mann, D., Boeve, B., Feldman, H., and Hutton, M. (2006) Mutations in progranulin cause tau-negative frontotemporal dementia linked to chromosome 17, Nature 442, 916-919. 108. Chu, T. T., Gao, N., Li, Q. Q., Chen, P. G., Yang, X. F., Chen, Y. X., Zhao, Y. F., and Li, Y. M. (2016) Specific Knockdown of Endogenous Tau Protein by Peptide-Directed Ubiquitin-Proteasome Degradation, Cell Chem Biol 23, 453-461. 109. Dorval, V., and Fraser, P. E. (2006) Small ubiquitin-like modifier (SUMO) modification of natively unfolded proteins tau and alpha-synuclein, J Biol Chem 281, 9919-9924. 110. Luo, H. B., Xia, Y. Y., Shu, X. J., Liu, Z. C., Feng, Y., Liu, X. H., Yu, G., Yin, G., Xiong, 30

ACS Paragon Plus Environment

Page 31 of 45 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

Y. S., Zeng, K., Jiang, J., Ye, K., Wang, X. C., and Wang, J. Z. (2014) SUMOylation at K340 inhibits tau degradation through deregulating its phosphorylation and ubiquitination, Proc Natl Acad Sci U S A 111, 16586-16591. 111. Takahashi, K., Ishida, M., Komano, H., and Takahashi, H. (2008) SUMO-1 immunoreactivity co-localizes with phospho-Tau in APP transgenic mice but not in mutant Tau transgenic mice, Neurosci Lett 441, 90-93. 112. Yan, S. D., Yan, S. F., Chen, X., Fu, J., Chen, M., Kuppusamy, P., Smith, M. A., Perry, G., Godman, G. C., Nawroth, P., and et al. (1995) Non-enzymatically glycated tau in Alzheimer's disease induces neuronal oxidant stress resulting in cytokine gene expression and release of amyloid beta-peptide, Nat Med 1, 693-699. 113. Ledesma, M. D., Bonay, P., Colaco, C., and Avila, J. (1994) Analysis of microtubule-associated protein tau glycation in paired helical filaments, J Biol Chem 269, 21614-21619. 114. Ledesma, M. D., Bonay, P., and Avila, J. (1995) Tau protein from Alzheimer's disease patients is glycated at its tubulin-binding domain, J Neurochem 65, 1658-1664. 115. Ko, L. W., Ko, E. C., Nacharaju, P., Liu, W. K., Chang, E., Kenessey, A., and Yen, S. H. (1999) An immunochemical study on tau glycation in paired helical filaments, Brain Res 830, 301-313. 116. Necula, M., and Kuret, J. (2004) Pseudophosphorylation and glycation of tau protein enhance but do not trigger fibrillization in vitro, J Biol Chem 279, 49694-49703. 117. Reyes, J. F., Geula, C., Vana, L., and Binder, L. I. (2012) Selective tau tyrosine nitration in non-AD tauopathies, Acta Neuropathol 123, 119-132. 118. Reyes, J. F., Fu, Y., Vana, L., Kanaan, N. M., and Binder, L. I. (2011) Tyrosine nitration within the proline-rich region of Tau in Alzheimer's disease, Am J Pathol 178, 2275-2285. 119. Reynolds, M. R., Berry, R. W., and Binder, L. I. (2005) Site-specific nitration differentially influences tau assembly in vitro, Biochemistry 44, 13997-14009. 120. Wang, Y., Garg, S., Mandelkow, E. M., and Mandelkow, E. (2010) Proteolytic processing of tau, Biochem Soc Trans 38, 955-961. 121. Wang, Y. P., Biernat, J., Pickhardt, M., Mandelkow, E., and Mandelkow, E. M. (2007) Stepwise proteolysis liberates tau fragments that nucleate the Alzheimer-like aggregation of full-length tau in a neuronal cell model, Proc Natl Acad Sci U S A 104, 10252-10257. 122. Zhang, Z., Song, M., Liu, X., Kang, S. S., Kwon, I. S., Duong, D. M., Seyfried, N. T., Hu, W. T., Liu, Z., Wang, J. Z., Cheng, L., Sun, Y. E., Yu, S. P., Levey, A. I., and Ye, K. (2014) Cleavage of tau by asparagine endopeptidase mediates the neurofibrillary pathology in Alzheimer's disease, Nat Med 20, 1254-1262. 123. Wischik, C. M., Novak, M., Thogersen, H. C., Edwards, P. C., Runswick, M. J., Jakes, R., Walker, J. E., Milstein, C., Roth, M., and Klug, A. (1988) Isolation of a fragment of tau derived from the core of the paired helical filament of Alzheimer disease, Proc Natl Acad Sci U S A 85, 4506-4510. 31

ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 32 of 45

124. de Calignon, A., Fox, L. M., Pitstick, R., Carlson, G. A., Bacskai, B. J., Spires-Jones, T. L., and Hyman, B. T. (2010) Caspase activation precedes and leads to tangles, Nature 464, 1201-1204. 125. Zhou, Y., Shi, J., Chu, D., Hu, W., Guan, Z., Gong, C. X., Iqbal, K., and Liu, F. (2018) Relevance of Phosphorylation and Truncation of Tau to the Etiopathogenesis of Alzheimer's Disease, Front Aging Neurosci 10, 27. 126. Kovacech, B., and Novak, M. (2010) Tau truncation is a productive posttranslational modification of neurofibrillary degeneration in Alzheimer's disease, Curr Alzheimer Res 7, 708-716. 127. Zilkova, M., Zilka, N., Kovac, A., Kovacech, B., Skrabana, R., Skrabanova, M., and Novak, M. (2011) Hyperphosphorylated truncated protein tau induces caspase-3 independent apoptosis-like pathway in the Alzheimer's disease cellular model, J Alzheimers Dis 23, 161-169. 128. Schweers, O., Schonbrunn-Hanebeck, E., Marx, A., and Mandelkow, E. (1994) Structural studies of tau protein and Alzheimer paired helical filaments show no evidence for beta-structure, J Biol Chem 269, 24290-24297. 129. von Bergen, M., Barghorn, S., Biernat, J., Mandelkow, E. M., and Mandelkow, E. (2005) Tau aggregation is driven by a transition from random coil to beta sheet structure, Biochim Biophys Acta 1739, 158-166. 130. Chirita, C. N., Congdon, E. E., Yin, H., and Kuret, J. (2005) Triggers of full-length tau aggregation: a role for partially folded intermediates, Biochemistry 44, 5862-5872. 131. Von, B. M., Friedhoff, P., Biernat, J., Heberle, J., Mandelkow, E. M., and Mandelkow, E. (2000) Assembly of tau protein into Alzheimer paired helical filaments depends on a local sequence motif ((306)VQIVYK(311)) forming beta structure, Proceedings of the National Academy of Sciences of the United States of America 97, 5129-5134. 132. Von, B. M., Barghorn, S., Li, L., Marx, A., Biernat, J., Mandelkow, E. M., and Mandelkow, E. (2001) Mutations of tau protein in frontotemporal dementia promote aggregation of paired helical filaments by enhancing local beta-structure, Journal of Biological Chemistry 276, 48165-48174. 133. Mirbaha, H., Chen, D., Morazova, O. A., Ruff, K. M., Sharma, A. M., Liu, X., Goodarzi, M., Pappu, R. V., Colby, D. W., Mirzaei, H., Joachimiak, L. A., and Diamond, M. I. (2018) Inert and seed-competent tau monomers suggest structural origins of aggregation, Elife 7. 134. Cowan, C. M., and Mudher, A. (2013) Are tau aggregates toxic or protective in tauopathies?, Front Neurol 4, 114. 135. Lee, V. M., Goedert, M., and Trojanowski, J. Q. (2001) Neurodegenerative tauopathies, Annu Rev Neurosci 24, 1121-1159. 136. Zilka, N., Filipcik, P., Koson, P., Fialova, L., Skrabana, R., Zilkova, M., Rolkova, G., Kontsekova, E., and Novak, M. (2006) Truncated tau from sporadic Alzheimer's disease suffices to drive neurofibrillary degeneration in vivo, FEBS Lett 580, 3582-3588. 32

ACS Paragon Plus Environment

Page 33 of 45 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

137. Perez, M., Valpuesta, J. M., Medina, M., Montejo de Garcini, E., and Avila, J. (1996) Polymerization of tau into filaments in the presence of heparin: the minimal sequence required for tau-tau interaction, J Neurochem 67, 1183-1190. 138. Cho, J. H., and Johnson, G. V. (2004) Glycogen synthase kinase 3 beta induces caspase-cleaved tau aggregation in situ, J Biol Chem 279, 54716-54723. 139. Wang, J. Z., Grundke-Iqbal, I., and Iqbal, K. (2007) Kinases and phosphatases and tau sites involved in Alzheimer neurofibrillary degeneration, Eur J Neurosci 25, 59-68. 140. Alonso Adel, C., Mederlyova, A., Novak, M., Grundke-Iqbal, I., and Iqbal, K. (2004) Promotion of hyperphosphorylation by frontotemporal dementia tau mutations, J Biol Chem 279, 34873-34881. 141. Guillozet-Bongaarts, A. L., Cahill, M. E., Cryns, V. L., Reynolds, M. R., Berry, R. W., and Binder, L. I. (2006) Pseudophosphorylation of tau at serine 422 inhibits caspase cleavage: in vitro evidence and implications for tangle formation in vivo, J Neurochem 97, 1005-1014. 142. Dickey, C. A., Kamal, A., Lundgren, K., Klosak, N., Bailey, R. M., Dunmore, J., Ash, P., Shoraka, S., Zlatkovic, J., Eckman, C. B., Patterson, C., Dickson, D. W., Nahman, N. S., Jr., Hutton, M., Burrows, F., and Petrucelli, L. (2007) The high-affinity HSP90-CHIP complex recognizes and selectively degrades phosphorylated tau client proteins, J Clin Invest 117, 648-658. 143. Watanabe, A., Hong, W. K., Dohmae, N., Takio, K., Morishima-Kawashima, M., and Ihara, Y. (2004) Molecular aging of tau: disulfide-independent aggregation and non-enzymatic degradation in vitro and in vivo, J Neurochem 90, 1302-1311. 144. Barghorn, S., Zheng-Fischhofer, Q., Ackmann, M., Biernat, J., von Bergen, M., Mandelkow, E. M., and Mandelkow, E. (2000) Structure, microtubule interactions, and paired helical filament aggregation by tau mutants of frontotemporal dementias, Biochemistry 39, 11714-11721. 145. Rizzu, P., Van Swieten, J. C., Joosse, M., Hasegawa, M., Stevens, M., Tibben, A., Niermeijer, M. F., Hillebrand, M., Ravid, R., Oostra, B. A., Goedert, M., van Duijn, C. M., and Heutink, P. (1999) High prevalence of mutations in the microtubule-associated protein tau in a population study of frontotemporal dementia in the Netherlands, Am J Hum Genet 64, 414-421. 146. Lu, M., and Kosik, K. S. (2001) Competition for microtubule-binding with dual expression of tau missense and splice isoforms, Mol Biol Cell 12, 171-184. 147. Adams, S. J., DeTure, M. A., McBride, M., Dickson, D. W., and Petrucelli, L. (2010) Three repeat isoforms of tau inhibit assembly of four repeat tau filaments, PLoS One 5, e10810. 148. Schoch, K. M., DeVos, S. L., Miller, R. L., Chun, S. J., Norrbom, M., Wozniak, D. F., Dawson, H. N., Bennett, C. F., Rigo, F., and Miller, T. M. (2016) Increased 4R-Tau Induces Pathological Changes in a Human-Tau Mouse Model, Neuron 90, 941-947. 149. Mudher, A., Colin, M., Dujardin, S., Medina, M., Dewachter, I., Alavi Naini, S. M., Mandelkow, E. M., Mandelkow, E., Buee, L., Goedert, M., and Brion, J. P. (2017) What is 33

ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 34 of 45

the evidence that tau pathology spreads through prion-like propagation?, Acta Neuropathol Commun 5, 99. 150. Goedert, M., Jakes, R., Spillantini, M. G., Hasegawa, M., Smith, M. J., and Crowther, R. A. (1996) Assembly of microtubule-associated protein tau into Alzheimer-like filaments induced by sulphated glycosaminoglycans, Nature 383, 550-553. 151. Giustiniani, J., Guillemeau, K., Dounane, O., Sardin, E., Huvent, I., Schmitt, A., Hamdane, M., Buee, L., Landrieu, I., Lippens, G., Baulieu, E. E., and Chambraud, B. (2015) The FK506-binding protein FKBP52 in vitro induces aggregation of truncated Tau forms with prion-like behavior, FASEB J 29, 3171-3181. 152. Wischik, C. M., Schelter, B. O., Wischik, D. J., Storey, J. M. D., and Harrington, C. R. (2018) Modeling Prion-Like Processing of Tau Protein in Alzheimer's Disease for Pharmaceutical Development, J Alzheimers Dis 62, 1287-1303. 153. Sanders, D. W., Kaufman, S. K., DeVos, S. L., Sharma, A. M., Mirbaha, H., Li, A., Barker, S. J., Foley, A. C., Thorpe, J. R., Serpell, L. C., Miller, T. M., Grinberg, L. T., Seeley, W. W., and Diamond, M. I. (2014) Distinct tau prion strains propagate in cells and mice and define different tauopathies, Neuron 82, 1271-1288. 154. Kfoury, N., Holmes, B. B., Jiang, H., Holtzman, D. M., and Diamond, M. I. (2012) Trans-cellular propagation of Tau aggregation by fibrillar species, J Biol Chem 287, 19440-19451. 155. Falcon, B., Noad, J., McMahon, H., Randow, F., and Goedert, M. (2018) Galectin-8-mediated selective autophagy protects against seeded tau aggregation, J Biol Chem 293, 2438-2451. 156. Holmes, B. B., DeVos, S. L., Kfoury, N., Li, M., Jacks, R., Yanamandra, K., Ouidja, M. O., Brodsky, F. M., Marasa, J., Bagchi, D. P., Kotzbauer, P. T., Miller, T. M., Papy-Garcia, D., and Diamond, M. I. (2013) Heparan sulfate proteoglycans mediate internalization and propagation of specific proteopathic seeds, Proc Natl Acad Sci U S A 110, E3138-3147. 157. Wu, J. W., Herman, M., Liu, L., Simoes, S., Acker, C. M., Figueroa, H., Steinberg, J. I., Margittai, M., Kayed, R., Zurzolo, C., Di Paolo, G., and Duff, K. E. (2013) Small misfolded Tau species are internalized via bulk endocytosis and anterogradely and retrogradely transported in neurons, J Biol Chem 288, 1856-1870. 158. Takahashi, M., Miyata, H., Kametani, F., Nonaka, T., Akiyama, H., Hisanaga, S., and Hasegawa, M. (2015) Extracellular association of APP and tau fibrils induces intracellular aggregate formation of tau, Acta Neuropathol 129, 895-907. 159. Ihara, Y., Morishima-Kawashima, M., and Nixon, R. (2012) The ubiquitin-proteasome system and the autophagic-lysosomal system in Alzheimer disease, Cold Spring Harb Perspect Med 2. 160. Flach, K., Hilbrich, I., Schiffmann, A., Gartner, U., Kruger, M., Leonhardt, M., Waschipky, H., Wick, L., Arendt, T., and Holzer, M. (2012) Tau oligomers impair artificial membrane integrity and cellular viability, J Biol Chem 287, 43223-43233. 34

ACS Paragon Plus Environment

Page 35 of 45 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

161. Fontaine, S. N., Zheng, D., Sabbagh, J. J., Martin, M. D., Chaput, D., Darling, A., Trotter, J. H., Stothert, A. R., Nordhues, B. A., Lussier, A., Baker, J., Shelton, L., Kahn, M., Blair, L. J., Stevens, S. M., Jr., and Dickey, C. A. (2016) DnaJ/Hsc70 chaperone complexes control the extracellular release of neurodegenerative-associated proteins, EMBO J 35, 1537-1549. 162. Saman, S., Kim, W., Raya, M., Visnick, Y., Miro, S., Jackson, B., McKee, A. C., Alvarez, V. E., Lee, N. C., and Hall, G. F. (2012) Exosome-associated tau is secreted in tauopathy models and is selectively phosphorylated in cerebrospinal fluid in early Alzheimer disease, J Biol Chem 287, 3842-3849. 163. DeVos, S. L., Corjuc, B. T., Oakley, D. H., Nobuhara, C. K., Bannon, R. N., Chase, A., Commins, C., Gonzalez, J. A., Dooley, P. M., Frosch, M. P., and Hyman, B. T. (2018) Synaptic Tau Seeding Precedes Tau Pathology in Human Alzheimer's Disease Brain, Front Neurosci 12, 267. 164. Liu, L., Drouet, V., Wu, J. W., Witter, M. P., Small, S. A., Clelland, C., and Duff, K. (2012) Trans-synaptic spread of tau pathology in vivo, PLoS One 7, e31302. 165. Katsinelos, T., Zeitler, M., Dimou, E., Karakatsani, A., Muller, H. M., Nachman, E., Steringer, J. P., Ruiz de Almodovar, C., Nickel, W., and Jahn, T. R. (2018) Unconventional Secretion Mediates the Trans-cellular Spreading of Tau, Cell Rep 23, 2039-2055. 166. Plouffe, V., Mohamed, N. V., Rivest-McGraw, J., Bertrand, J., Lauzon, M., and Leclerc, N. (2012) Hyperphosphorylation and cleavage at D421 enhance tau secretion, PLoS One 7, e36873. 167. Wu, J. W., Hussaini, S. A., Bastille, I. M., Rodriguez, G. A., Mrejeru, A., Rilett, K., Sanders, D. W., Cook, C., Fu, H., Boonen, R. A., Herman, M., Nahmani, E., Emrani, S., Figueroa, Y. H., Diamond, M. I., Clelland, C. L., Wray, S., and Duff, K. E. (2016) Neuronal activity enhances tau propagation and tau pathology in vivo, Nat Neurosci 19, 1085-1092. 168. Bolos, M., Llorens-Martin, M., Jurado-Arjona, J., Hernandez, F., Rabano, A., and Avila, J. (2016) Direct Evidence of Internalization of Tau by Microglia In Vitro and In Vivo, J Alzheimers Dis 50, 77-87. 169. Maphis, N., Xu, G., Kokiko-Cochran, O. N., Jiang, S., Cardona, A., Ransohoff, R. M., Lamb, B. T., and Bhaskar, K. (2015) Reactive microglia drive tau pathology and contribute to the spreading of pathological tau in the brain, Brain 138, 1738-1755. 170. Asai, H., Ikezu, S., Tsunoda, S., Medalla, M., Luebke, J., Haydar, T., Wolozin, B., Butovsky, O., Kugler, S., and Ikezu, T. (2015) Depletion of microglia and inhibition of exosome synthesis halt tau propagation, Nat Neurosci 18, 1584-1593. 171. Ke, Y. D., Suchowerska, A. K., van der Hoven, J., De Silva, D. M., Wu, C. W., van Eersel, J., Ittner, A., and Ittner, L. M. (2012) Lessons from tau-deficient mice, Int J Alzheimers Dis 2012, 873270. 172. Harada, A., Oguchi, K., Okabe, S., Kuno, J., Terada, S., Ohshima, T., Sato-Yoshitake, R., Takei, Y., Noda, T., and Hirokawa, N. (1994) Altered microtubule organization in small-calibre axons of mice lacking tau protein, Nature 369, 488-491. 35

ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 36 of 45

173. Roberson, E. D., Scearce-Levie, K., Palop, J. J., Yan, F., Cheng, I. H., Wu, T., Gerstein, H., Yu, G. Q., and Mucke, L. (2007) Reducing endogenous tau ameliorates amyloid beta-induced deficits in an Alzheimer's disease mouse model, Science 316, 750-754. 174. DeVos, S. L., Goncharoff, D. K., Chen, G., Kebodeaux, C. S., Yamada, K., Stewart, F. R., Schuler, D. R., Maloney, S. E., Wozniak, D. F., Rigo, F., Bennett, C. F., Cirrito, J. R., Holtzman, D. M., and Miller, T. M. (2013) Antisense reduction of tau in adult mice protects against seizures, J Neurosci 33, 12887-12897. 175. Santa-Maria, I., Alaniz, M. E., Renwick, N., Cela, C., Fulga, T. A., Van Vactor, D., Tuschl, T., Clark, L. N., Shelanski, M. L., McCabe, B. D., and Crary, J. F. (2015) Dysregulation of microRNA-219 promotes neurodegeneration through post-transcriptional regulation of tau, J Clin Invest 125, 681-686. 176. Banzhaf-Strathmann, J., Benito, E., May, S., Arzberger, T., Tahirovic, S., Kretzschmar, H., Fischer, A., and Edbauer, D. (2014) MicroRNA-125b induces tau hyperphosphorylation and cognitive deficits in Alzheimer's disease, EMBO J 33, 1667-1680. 177. Smith, P. Y., Hernandez-Rapp, J., Jolivette, F., Lecours, C., Bisht, K., Goupil, C., Dorval, V., Parsi, S., Morin, F., Planel, E., Bennett, D. A., Fernandez-Gomez, F. J., Sergeant, N., Buee, L., Tremblay, M. E., Calon, F., and Hebert, S. S. (2015) miR-132/212 deficiency impairs tau metabolism and promotes pathological aggregation in vivo, Hum Mol Genet 24, 6721-6735. 178. Liu, W., Zhao, J., and Lu, G. (2016) miR-106b inhibits tau phosphorylation at Tyr18 by targeting Fyn in a model of Alzheimer's disease, Biochem Biophys Res Commun 478, 852-857. 179. Wang, Y., Yang, R., Gu, J., Yin, X., Jin, N., Xie, S., Chang, H., Qian, W., Shi, J., Iqbal, K., Gong, C. X., Cheng, C., and Liu, F. (2015) Cross talk between PI3K-AKT-GSK-3beta and PP2A pathways determines tau hyperphosphorylation, Neurobiol Aging 36, 188-200. 180. Hochgrafe, K., Sydow, A., Matenia, D., Cadinu, D., Konen, S., Petrova, O., Pickhardt, M., Goll, P., Morellini, F., Mandelkow, E., and Mandelkow, E. M. (2015) Preventive methylene blue treatment preserves cognition in mice expressing full-length pro-aggregant human Tau, Acta Neuropathol Commun 3, 25. 181. Levenson, J. M., Schroeter, S., Carroll, J. C., Cullen, V., Asp, E., Proschitsky, M., Chung, C. H., Gilead, S., Nadeem, M., Dodiya, H. B., Shoaga, S., Mufson, E. J., Tsubery, H., Krishnan, R., Wright, J., Solomon, B., Fisher, R., and Gannon, K. S. (2016) NPT088 reduces both amyloid-beta and tau pathologies in transgenic mice, Alzheimers Dement (N Y) 2, 141-155. 182. Seidler, P. M., Boyer, D. R., Rodriguez, J. A., Sawaya, M. R., Cascio, D., Murray, K., Gonen, T., and Eisenberg, D. S. (2018) Structure-based inhibitors of tau aggregation, Nat Chem 10, 170-176. 183. Cao, Y. N., Zheng, L. L., Wang, D., Liang, X. X., Gao, F., and Zhou, X. L. (2018) Recent advances in microtubule-stabilizing agents, Eur J Med Chem 143, 806-828. 184. Winblad, B., Graf, A., Riviere, M. E., Andreasen, N., and Ryan, J. M. (2014) Active 36

ACS Paragon Plus Environment

Page 37 of 45 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

immunotherapy options for Alzheimer's disease, Alzheimers Res Ther 6, 7. 185. Kontsekova, E., Zilka, N., Kovacech, B., Novak, P., and Novak, M. (2014) First-in-man tau vaccine targeting structural determinants essential for pathological tau-tau interaction reduces tau oligomerisation and neurofibrillary degeneration in an Alzheimer's disease model, Alzheimers Res Ther 6, 44. 186. Novak, P., Schmidt, R., Kontsekova, E., Zilka, N., Kovacech, B., Skrabana, R., Vince-Kazmerova, Z., Katina, S., Fialova, L., Prcina, M., Parrak, V., Dal-Bianco, P., Brunner, M., Staffen, W., Rainer, M., Ondrus, M., Ropele, S., Smisek, M., Sivak, R., Winblad, B., and Novak, M. (2017) Safety and immunogenicity of the tau vaccine AADvac1 in patients with Alzheimer's disease: a randomised, double-blind, placebo-controlled, phase 1 trial, Lancet Neurol 16, 123-134. 187. Theunis, C., Crespo-Biel, N., Gafner, V., Pihlgren, M., Lopez-Deber, M. P., Reis, P., Hickman, D. T., Adolfsson, O., Chuard, N., Ndao, D. M., Borghgraef, P., Devijver, H., Van Leuven, F., Pfeifer, A., and Muhs, A. (2013) Efficacy and safety of a liposome-based vaccine against protein Tau, assessed in tau.P301L mice that model tauopathy, PLoS One 8, e72301. 188. Doody, R. (2017) Developing Disease-Modifying Treatments in Alzheimer's Disease - A Perspective from Roche and Genentech, J Prev Alzheimers Dis 4, 264-272. 189. Castillo-Carranza, D. L., Sengupta, U., Guerrero-Munoz, M. J., Lasagna-Reeves, C. A., Gerson, J. E., Singh, G., Estes, D. M., Barrett, A. D., Dineley, K. T., Jackson, G. R., and Kayed, R. (2014) Passive immunization with Tau oligomer monoclonal antibody reverses tauopathy phenotypes without affecting hyperphosphorylated neurofibrillary tangles, J Neurosci 34, 4260-4272. 190. Castillo-Carranza, D. L., Gerson, J. E., Sengupta, U., Guerrero-Munoz, M. J., Lasagna-Reeves, C. A., and Kayed, R. (2014) Specific targeting of tau oligomers in Htau mice prevents cognitive impairment and tau toxicity following injection with brain-derived tau oligomeric seeds, J Alzheimers Dis 40 Suppl 1, S97-S111. 191. Panza, F., Solfrizzi, V., Seripa, D., Imbimbo, B. P., Lozupone, M., Santamato, A., Tortelli, R., Galizia, I., Prete, C., Daniele, A., Pilotto, A., Greco, A., and Logroscino, G. (2016) Tau-based therapeutics for Alzheimer's disease: active and passive immunotherapy, Immunotherapy 8, 1119-1134. 192. Graham, W. V., Bonito-Oliva, A., and Sakmar, T. P. (2017) Update on Alzheimer's Disease Therapy and Prevention Strategies, Annu Rev Med 68, 413-430. 193. Gallardo, G., and Holtzman, D. M. (2017) Antibody Therapeutics Targeting Abeta and Tau, Cold Spring Harb Perspect Med 7. 194. Dai, C. L., Chen, X., Kazim, S. F., Liu, F., Gong, C. X., Grundke-Iqbal, I., and Iqbal, K. (2015) Passive immunization targeting the N-terminal projection domain of tau decreases tau pathology and improves cognition in a transgenic mouse model of Alzheimer disease and tauopathies, J Neural Transm (Vienna) 122, 607-617. 195. Dai, C. L., Hu, W., Tung, Y. C., Liu, F., Gong, C. X., and Iqbal, K. (2018) Tau passive 37

ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 38 of 45

immunization blocks seeding and spread of Alzheimer hyperphosphorylated Tau-induced pathology in 3 x Tg-AD mice, Alzheimers Res Ther 10, 13. 196. Dai, C. L., Tung, Y. C., Liu, F., Gong, C. X., and Iqbal, K. (2017) Tau passive immunization inhibits not only tau but also Abeta pathology, Alzheimers Res Ther 9, 1. 197. Bittar, A., Sengupta, U., and Kayed, R. (2018) Prospects for strain-specific immunotherapy in Alzheimer's disease and tauopathies, NPJ Vaccines 3, 9. 198. Guerrero-Munoz, M. J., Gerson, J., and Castillo-Carranza, D. L. (2015) Tau Oligomers: The Toxic Player at Synapses in Alzheimer's Disease, Front Cell Neurosci 9, 464. 199. Usenovic, M., Niroomand, S., Drolet, R. E., Yao, L., Gaspar, R. C., Hatcher, N. G., Schachter, J., Renger, J. J., and Parmentier-Batteur, S. (2015) Internalized Tau Oligomers Cause Neurodegeneration by Inducing Accumulation of Pathogenic Tau in Human Neurons Derived from Induced Pluripotent Stem Cells, J Neurosci 35, 14234-14250. 200. Gerson, J. E., Sengupta, U., and Kayed, R. (2017) Tau Oligomers as Pathogenic Seeds: Preparation and Propagation In Vitro and In Vivo, Methods Mol Biol 1523, 141-157. 201. Wang, J. Z., and Liu, F. (2008) Microtubule-associated protein tau in development, degeneration and protection of neurons, Prog Neurobiol 85, 148-175. 202. Yuzwa, S. A., Yadav, A. K., Skorobogatko, Y., Clark, T., Vosseller, K., and Vocadlo, D. J. (2011) Mapping O-GlcNAc modification sites on tau and generation of a site-specific O-GlcNAc tau antibody, Amino Acids 40, 857-868. 203. Smet-Nocca, C., Broncel, M., Wieruszeski, J. M., Tokarski, C., Hanoulle, X., Leroy, A., Landrieu, I., Rolando, C., Lippens, G., and Hackenberger, C. P. (2011) Identification of O-GlcNAc sites within peptides of the Tau protein and their impact on phosphorylation, Mol Biosyst 7, 1420-1429.

38

ACS Paragon Plus Environment

Page 39 of 45 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

Figure 1: Gene expression of human MAPT and protein structure of tau isoforms. Human MAPT gene locates on chromosome 17 and comprises 16 exons. Exon 0 and exon 1 encode the 5' untranslated region of MAPT mRNA, while exon 14 is transcribed into part of the 3' untranslated region. Exons 4a, 6, and 8 are expressed only in peripheral tissues. Alternative splicing of exons 2 (green, encodes N1) and 3 (yellow, encodes N2) produces tau isoforms with zero (0N), one (1N) or two (2N) N-terminal inserts of 29-amino acids each. Exons 9-12 encode microtubule-binding repeats (R1, R3 and R4 in blue, R2 in red). Alternative splicing of exon 10 (red, encodes R2) produces isoforms containing four (4R-tau) or three (3R-tau) microtubule-binding repeats.

39

ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 40 of 45

Figure 2. Structure and function of physiological and pathological tau. (A) Tau is natively unfolded (inert monomer, blue) in normal neurons. The major function of tau is to stabilize microtubules through the microtubule-binding domain (blue boxes). (B) In the diseased neurons from AD brain, post-translational modifications of tau, especially truncation and hyperphosphorylation, facilitate the detachment of pathological tau from microtubules, resulting in depolymerization of microtubules. Conformational change of tau converts the inert monomer into a seed-competent form (red), which may template the aggregation of larger assemblies, including dimers, oligomers, and finally PHFs. (C) Seed-competent monomers and aggregates transport from one neuron to another via multiple pathways. Internalized tau seeds could be eliminated through endosomal-lysosomal or Galectin-8-mediated (green hexagon) autophagic system. Once escaping from the damaged endosomal vesicles, tau seeds sequester cytoplasmic inert tau and template the formation of new seeds in a prion-like mechanism. (D) Tau seeds spread along synaptically connected neurons to affect large-scale brain networks, leading to the stereotypical propagation of tau pathology.

40

ACS Paragon Plus Environment

Page 41 of 45 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

41

ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 42 of 45

Table of Contents Graphic

Manuscript title: Pathological Changes of Tau Related to Alzheimer’s Disease Authors: Dandan Chu and Fei Liu

For Table of Contents Use Only

42

ACS Paragon Plus Environment

Page 43 of 45 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

Figure 1 99x58mm (300 x 300 DPI)

ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Figure 2 141x152mm (300 x 300 DPI)

ACS Paragon Plus Environment

Page 44 of 45

Page 45 of 45 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

Table of contents graphic 95x34mm (300 x 300 DPI)

ACS Paragon Plus Environment