Download as pdf or txt
Download as pdf or txt
You are on page 1of 23

Journal of Alzheimer’s Disease 1 (1999) 329–351 329

IOS Press

Tau Protein in Normal and Alzheimer’s Disease


Brain: An Update*
Gail V.W. Johnson** and Judith A. Hartigan KEYWORDS: Microtubule, phosphorylation, PHF,
kinase, phosphatase
Department of Psychiatry and Behavioral
Neurobiology, University of Alabama at INTRODUCTION
Birmingham, Birmingham, AL 35294-0017,
USA Tau is a microtubule-associated protein that is
selectively, but not exclusively (3,159),
expressed in the nervous system. In addition,
ABSTRACT: Tau is a microtubule-associated protein there is evidence of evolutionary conservation, as
that, in a hyperphosphorylated form, comprises the tau-like proteins have been identified in
main component of the paired helical filaments and
Caenorhabditis elegans (102). In Alzheimer’s
neurofibrillary tangles found in Alzheimer’s Disease
(AD) brain. It is therefore important to understand the
Disease (AD) brain tau is abnormally phos-
normal functioning and processing of tau protein, and phorylated. This hyperphosphorylated form of
the abnormal posttranslational processing of tau in AD tau is the major component of the paired helical
pathology. In 1996, Johnson and Jenkins reviewed the filaments (PHFs) and neurofibrillary tangles
literature on the biochemistry, function, and phos- (NFTs) found in the AD brain. The mechanisms
phorylation of tau in normal and AD brain. Since that underlying the abnormal phosphorylation and
time, numerous publications have come out further accumulation of tau in AD are still under
elucidating the properties of tau. The present review investigation.
updates the topics originally covered in the 1996 In 1996, Johnson and Jenkins (74) reviewed
review, as well as presents a number of new topics.
the biochemistry, phosphorylation and function
For example, mutations in the tau gene have been
found in several non-AD, autosomal dominant
of the tau protein in the normal and AD brain.
neurodegenerative disorders that exhibit extensive However, since that time, a significant number of
neurofibrillary pathology. In addition, there is articles have been published further elucidating
increasing evidence that tau may be involved in signal the properties of tau. This review will present an
transduction, organelle transport, and cell growth, update on tau in normal and AD brain since the
independent of its microtubule-binding functions. time of the 1996 Johnson and Jenkins publication
Taken together, the research reviewed here (74). In addition to presenting new information
demonstrates that tau is a very complex protein with on the topics originally covered in the 1996
various functions that are intricately regulated. It is review, several new areas of research will be
clear that more research is required to completely
discussed. For example, mutations in the tau
understand the functions and regulation of tau in
normal and AD brain.
gene have been found in several families with
autosomal dominant forms of presenile non-AD
dementia. Also, there has been an explosion of
*
This article is published with permission from the University articles published on the role of glycogen
of Kentucky. It is also published in the online journal
synthase kinase 3β (GSK-3β) in modulating tau
Alzheimer’s Disease Review; www.coa.uky.edu/ADReview/
phosphorylation, and this work will be discussed.
**
Corresponding author: Dr. Gail V.W. Johnson, The presence of tau in the cerebral spinal fluid
Department of Psychiatry, SC 1061, 1720 7th Avenue South, (CSF), which may be useful in the diagnosis of
University of Alabama at Birmingham, Birmingham, AL AD, will also be reviewed. Finally, there is new
35294-0017, USA, E-mail: gvwj@uab.edu

© University of Kentucky
330 G.V.W. Johnson and J.A. Hartigan / Tau Protein in Normal and Alzheimer’s Disease Brain

evidence to suggest some novel roles for tau, (47). Using optical tweezers to bend individual
such as being involved in signal transduction and microtubules, Felgner (33) measured flexural
axonal outgrowth in a non-microtubule- rigidity by their return movement to the resting
associated manner. position in the presence of various tau constructs.
When full-length tau was bound, the flexural
rigidity of the microtubules was increased almost
3-fold over that observed with pure microtubules.
TAU AND PHF-tau: Tau constructs containing only the repeat regions
STRUCTURE/FUNCTION were not efficient at increasing the rigidity of the
microtubules, however addition of flanking
Historically, the C-terminal repeat regions of domains to the tau constructs greatly enhanced
the tau molecule were defined as microtubule their ability to decrease the flexibility of the
binding repeats (see Fig. 1). However, recent microtubules (33). In situ evidence of the role
studies have revealed that both the repeat regions these different domains of tau play in modulating
and the flanking domains are required for tau-microtubule interactions was obtained by
efficient and productive tau-microtubule inter- injecting different tau isoforms and constructs
actions. In vitro, tau constructs containing only into Chinese hamster ovary (CHO) cells, then
the repeat regions of tau bound microtubules with analyzing the association of tau with micro-
a relatively low affinity, however this region tubules (123). The results showed that the
efficiently promoted microtubule assembly and presence of both the flanking regions and the
stability. In contrast, the flanking domains of tau repeat domains was necessary for tau to
bound microtubules with a relatively high efficiently interact with microtubules in the cells,
affinity, but did not effectively enhance micro- further supporting the hypothesis that the
tubule stability (49). These findings indicate that flanking and repeat domains play important
the “targetting” flanking domains of tau are likely cooperative roles in modulating tau function
responsible for positioning tau on the micro- (123).
tubule surface, so that the “functional” repeat Taken together, these studies clearly indicate
domains can efficiently promote microtubule that the regions of tau that have been referred to
stability and assembly. This model stresses the as the “microtubule-binding domains” are the
importance of both the flanking and the repeat important functional domains in terms of
domains for optimal tau-microtubule interactions. promoting microtubule assembly and stability.
In further support of this hypothesis, deletion of However, the flanking regions are essential for
the amino third of tau did not significantly alter targetting tau to the microtubules and contribute
tau-microtubule interactions, however deletion of to the function of tau as a microtubule stabilizing
∼ 30 amino acids within the proline-rich amino protein (123). In addition, the flanking domains
terminal flanking domain drastically reduced the of tau are hyperphosphorylated in AD brain, and
affinity of the tau construct for microtubules this likely contributes significantly to the

Fig. 1. Schematic Representation of the Tau Protein and the Location of PHF-Tau Phosphorylation Sites. Sites
phosphorylated in PHF-tau (107,176) are included on the top of the molecule, with Ser-Thr/Pro sites designated by
asterisks (*). Diagram is based on the 441 residue isoform (43). Flanking regions are based on (49).
G.V.W. Johnson and J.A. Hartigan / Tau Protein in Normal and Alzheimer’s Disease Brain 331

microtubule-binding incompetence of AD tau (numbering according to the longest human brain


(107). tau isoform (43)) within the repeat domain of tau
A likely outcome of alterations in tau (76). This cysteine was previously shown to play
phosphorylation is changes in the conformational a critically important role in the in vitro assembly
state of tau. Protein kinase A phosphorylation of of different tau constructs into PHF-like
two serine residues in the flanking domains structures (134). Free fatty acids have been
causes a shift in electrophoretic mobility which is shown to enhance filament formation of tau,
indicative of a conformational change (16). although the filaments were not PHF-like in
Further, mutation of these serine residues to structure (170). Tissue transglutaminase (Tgase)
aspartate residues to mimic phosphorylation is a calcium-dependent cross-linking enzyme that
results in conformational changes similar to those has been proposed to play a role in the function
caused by phosphorylation (89). In addition, it of the insoluble NFTs in AD brain (31,138).
has been demonstrated that the antibody Alz-50 Tgase modifies human recombinant tau in vitro
recognizes tau in a specific conformation, and it resulting in the formation of insoluble
has been postulated that aberrant tau phos- filamentous structures (4). Further, these fila-
phorylation and/or polymerization into PHFs mentous structures stained with an antibody to
induces the conformation that is recognized Tgase just as strongly as PHFs isolated from AD
selectively by Alz-50 (21). In vitro, PHF-tau brain stained with the same antibody, suggesting
binds normally phosphorylated tau forming that cross-linking of tau by Tgase may be
filamentous aggregates, suggesting hyper- involved in AD pathology (4). The self-
phosphorylated tau may be able to facilitate PHF aggregation of tau into filaments can be inhibited
formation by sequestering normal tau (2). by the phenothiazine, methylene blue, and further
Although phosphorylation of specific sites it reversed the proteolytic-resistance of PHFs by
may facilitate the aggregation of tau into interrupting the tau-tau interactions. This effect
filaments in AD brain, it is likely that other of methylene blue was reportedly due to its
factors contribute to this process. For example, binding to the repeat region of tau thereby
incubation of non-phosphorylated tau with inhibiting tau-tau interactions (171).
sulfated glycosaminoglycans, such as heparin or Recently it has been hypothesized that
heparan sulfate, resulted in the formation of PHF- differential expression of specific exons may
like structures (8,41,53,119). In addition, the affect the ability of tau to form PHFs and NFTs.
repeat region of tau was essential for the Nelson et al. (113) cloned and sequenced tau-
induction of PHF formation (41). Heparin also encoding mRNA transcripts from the brains of
prevented tau from binding to microtubules and rhesus monkeys (which do not develop NFTs)
promoted microtubule disassembly. Additionally, and of domesticated goat (which do develop
there is some evidence to suggest that heparan NFTs). The deduced amino acid sequence of
sulfate and tau may co-localize in NFT- human tau has been previously reported (42,43).
containing neurons in AD brain (41,78,120, The main difference between monkey, human
144,145). Considering this, Goedert et al. (41) and goat tau mRNA transcripts was the presence
hypothesized that “an increase in sulphated of exon 8 in monkeys. This exon was not found
glycosaminoglycans within the cytoplasm of in humans or goat. Exon 8 includes a highly
nerve cells may trigger the hyperphosphorylation conserved Pro-Pro-Pro motif that would be
of tau, destabilize microtubules and induce expected to introduce a sharp bend into the
assembly of PHFs”. However, further research is protein structure. It was speculated that this bend
required to elucidate the potential role of sulfated may sterically hinder phosphorylation and
glycosaminoglycans in AD pathology. polymerization, making animals that possess
RNA also strongly enhances PHF assembly exon 8 less susceptible to NFT formation (113).
and requires the formation of intermolecular In an earlier study using a polyclonal antibody to
disulfide bridges involving the Cys322 residue exon 8, no exon 8 was detected in human tau
332 G.V.W. Johnson and J.A. Hartigan / Tau Protein in Normal and Alzheimer’s Disease Brain

(22). However, there was also no exon 8 present event that coincided with increased tau
in rat or rabbit tau, which are species that do not phosphorylation. Thus it was suggested that in
develop NFTs (22). Taken together, these data axons, regulation of tau phosphorylation may
indicate that the purported role of exon 8 in NFT modulate motor protein-dependent axonal
formation requires further study before any transport (129).
conclusions can be made. There is a growing body of evidence that tau
interacts either directly or indirectly with the
actin cytoskeleton and thereby plays a role in
NOVEL FUNCTIONS regulating cell shape, motility and microtubule-
plasma membrane interactions. The M2 cell line,
Tau is a microtubule-associated protein whose derived from a malignant melanoma, lacks the
historical role has been to promote microtubule major actin-binding protein, ABP-280 (filamin),
stability and assembly. However, recently a and exhibits extensive membrane blebbing.
number of papers have been published describing Microinjections of tau or MAP2 prevented bleb
functions of tau beyond its well-characterized formation and restored a normal phenotype (26).
ability to modulate microtubule dynamics. Here Suppression of tau expression in cultured neurons
we will describe several of the novel functions using antisense oligonucleotides results in a
recently attributed to tau. significant reduction in growth cone area and
In 1996, Hwang et al. (63) demonstrated that fillopodia number concomitant with considerable
tau interacted with a key enzyme in the changes in the phallodin staining of F-actin
phosphatidylinositol signalling pathway, phos- (29). In addition, localization of tau to the distal
pholipase C-γ (PLC-γ). The authors found that in axon requires intact microfilaments (79). Al-
vitro, PLC-γ was maximally activated by a though these data indicate that tau may interact
combination of tau and arachidonic acid (63). with actin directly, it is more likely that tau is
Further it was demonstrated that in human interacting with actin through other proteins, as
neuroblastoma cells, tau selectively associates in vitro MAP-2C efficiently promotes actin
with PLC-γ, and the PLC-γ that co-precipitates gelatination, but tau does not (26). Additionally,
with tau is enzymatically active (69). Ad- tau does not show a direct co-localization with
ditionally, in vitro tau acts as a phosphoti- the microfilament network, indicating the exis-
dylinositol bisphosphate (PIP2) binding protein tence of binding partners which could localize tau
like gelsolin, and it was suggested that the direct to the actin cytoskeleton (79).
binding of tau to PIP2 may make PIP2 a more Neurite outgrowth is a specialized form of cell
favorable substrate for PLC-γ (34). These data motility, and it is clear that tau plays a role in this
strongly indicate that tau is likely to play a role in process. Growing axons are very dynamic
signal transduction. structures and the stability of the microtubules
In addition to modulating the stability of varies along the length of the axon. The distal
microtubules, tau is likely to regulate the motor- end of the axon is more sensitive to microtubule
protein mediated transport of organelles along depolymerizing drugs and the tubulin turnover is
microtubules. Transfection of tau into COS cells, more rapid than in the proximal end. Con-
which do not normally express tau, resulted in a sidering the role of tau as a microtubule
significant decrease in motor-based organelle stabilizing protein, it was hypothesized that tau
transport (129). In vitro, tau binds to one heavy would be more concentrated at the more stable,
chain of the motor protein kinesin and the proximal end and less concentrated at the more
presence of tau decreases the ability of micro- dynamic, distal end. When this was tested in
tubules to stimulate the MgATPase activity of cultured rat sympathetic neurons, Black et al.
purified kinesin (68). Additionally, activation of (13) found the exact opposite. That is, tau was
protein kinase A in cultured sensory neurons most concentrated at the distal end of the axon, in
increased the transport of large organelles, an the growth cones (13). Similar results were
G.V.W. Johnson and J.A. Hartigan / Tau Protein in Normal and Alzheimer’s Disease Brain 333

obtained using cultured rat hippocampal neurons apparently the most efficient at phosphorylating
(79). Since in the distal regions of the growing tau in vitro (39,125,126). Based on immunoblot
axon the microtubules are most dynamic, this analysis using antibodies against specific
data strongly suggests that tau in the growing phospho-epitopes, the SAPKs have been shown
axon has functions other than increasing to phosphorylate several of the sites on tau that
microtubule stability. One possibility is that the are phosphorylated in AD brain (39,125,126).
tau in the developing neuron contains only three Another proline-directed protein kinase that
repeat regions, since tau is developmentally phosphorylates tau and has received significant
regulated, and this isoform has a reduced ability attention lately is glycogen synthase kinase 3β
to bind and stabilize microtubules. (GSK-3β, also called tau protein kinase I
In addition to concentration changes in tau (TPKI)). Earlier work demonstrated that tau is a
protein in the developing neuron, the phos- substrate of GSK-3β (51,93) and that prior
phorylation state of tau was also found to differ phosphorylation of tau by cdk5 (TPKII)
along the length of the growing axon (95). Using enhanced the rate and extent of subsequent
fetal hippocampal cells, it was found that tau in phosphorylation by GSK-3β (66,139). Speci-
the growth cones was highly dephosphorylated at fically, phosphorylation at Thr231 by GSK-3β was
the Tau-1 site as compared to the somatodendritic enhanced 9-fold, which is a site that is hyper-
compartment. A phosphorylation gradient was phosphorylated in PHF-tau. Additionally, phos-
evident, with a gradual change from phos- phorylation of tau by GSK-3β has been shown to
phorylated to dephosphorylated tau from the reduce microtubule polymerization in vitro (161).
soma, through the axon, to the growth cone.
These studies again suggest that the function of
tau in the growing axon extends beyond its IN SITU PHOSPHORYLATION/DE-
classical microtubule stabilizing function because PHOSPHORYLATION STUDIES
in most cases dephosphorylation of tau increases
its affinity for microtubules. Recently numerous articles have been
published on the role of GSK-3β in tau phos-
phorylation in situ. Tau transiently expressed in
IN VITRO PHOSPHORYLATION non-neuronal mammalian cell lines is not
STUDIES phosphorylated significantly at sites found to be
phosphorylated in PHF-tau. However, when
In vitro, tau is phosphorylated by numerous GSK-3β is co-expressed with tau in these cells,
protein kinases (for earlier review see (75)), and numerous Ser/Thr-Pro sites that are found in
the list continues to grow (e.g., (117,172)). In PHF-tau become phosphorylated (65,92). Co-
Table 1, sites on tau that have been reported to be expression of tau with GSK-3β also resulted in
phosphorylated by specific protein kinases in less stable microtubules than when the cells were
vitro are listed. It should be noted that although transfected with only tau. Analogously, Wagner
many of the protein kinases and tau phosphory- et al. (167) transfected 3T3 and CHO cells with
lation sites are indicated, this is not a tau and GSK-3β. Cells transfected with tau alone
comprehensive list. Recently, members of the displayed prominent bundles of microtubules.
stress-activated protein kinase (SAPK) family However, co-transfection with tau and GSK-3β
have been shown to phosphorylate tau in vitro. led to increased tau phosphorylation and to a
SAPK1γ (or Jun N-terminal kinase, JNK1), decrease in microtubule bundling. The authors
SAPK2a (also known as p38, RK, CSBPs, Mpk2 also used a mutant form of tau in which five of
or Mxi2), SAPK2b (or p38β), SAPK3 (also the sites known to be phosphorylated by GSK-3β
known as ERK6 or p38γ) and SAPK4 all were converted to alanine to preclude
phosphorylate tau at multiple Ser/Thr-Pro phosphorylation. Transfection of this mutant
epitopes, although SAPK3 and SAPK4 are form of tau alone resulted in microtubule
334 G.V.W. Johnson and J.A. Hartigan / Tau Protein in Normal and Alzheimer’s Disease Brain

Table 1
Reported In Vitro Phosphorylation Sites on Tau by Various Protein Kinases. Asterisks (*) represent sites on tau that
are phosphorylated in PHF-tau and by various protein kinases. The references cited in the table are as follows:
a, (107); b, (75); c, (85); d, (64); e, (117); f, (126); g, (125); h, (39)

bundling as did co-transfection of GSK-3β with has been shown to be due to activation of
the mutant tau isoform (167). These results phosphotidylinositol-3-kinase (PI-3-K). PI-3-K
suggest that the phosphorylation of tau by GSK- activates Akt (also known as protein kinase B)
3β reduces tau’s ability to organize microtubules. which phosphorylates GSK-3β on the Ser9
Finally, Brownlees et al. (17) created transgenic residue resulting in inactivation (25,60). Recent-
mice which expressed either wild-type GSK-3β ly, insulin and IGF-1 were shown to decrease tau
or a mutant form of GSK-3β that was more phosphorylation in NT2N cells through a PI-3-
active, and examined the phosphorylation state of K — Akt — GSK-3β dependent pathway (60).
tau in the brains of the transgenic mice and non- These data strongly indicate that tau is a
transgenic controls. The transgenics with the physiological substrate of GSK-3β.
highest level of GSK-3β activity showed elevated In 1996, two reports clearly demonstrated that
tau phosphorylation using immunoblot analysis in vitro lithium is an uncompetitive, selective
with the AT8 antibody which recognizes tau inhibitor of GSK-3β (81,151). In cultured NT2N
when Ser202 and Thr205 are phosphorylated (17). cells, lithium inhibited GSK-3β activity, and this
Insulin and insulin growth factor-1 (IGF-1) inhibition resulted in a decrease in tau
have been shown to modulate the activity of phosphorylation (59). Further, these investi-
GSK-3β (60). Prolonged activation of the insulin gators found that lithium treatment reduced the
or IGF-1 receptors often results in a down- amount of tau that was associated with micro-
regulation of GSK-3β (60). In some systems this tubules. In SH-SY5Y cells, lithium treatment
G.V.W. Johnson and J.A. Hartigan / Tau Protein in Normal and Alzheimer’s Disease Brain 335

reduced the phosphorylation of tau at some, but phorylation of tau through two mechanisms.
not all, of the sites that were affected in NT2N Because tau is a substrate of PP2A (103,147),
cells. However, in contrast to what was observed decreased PP2A activity could directly result in
in NT2N cells, lithium treatment did not alter the increased tau phosphorylation. In addition, a
amount of tau that was associated with decrease in PP2A activity could lead to increased
microtubules (174). Lithium also decreased tau MARK activity, which may result in increased
phosphorylation in cultured rat neurons (109). Ser262 phosphorylation and lead to disruption of
Although GSK-3β is likely to be involved in microtubule assemblies.
modulating tau phosphorylation, the data sugges- There has also been a recent study that
ting a role for this kinase in the pathological indicates phosphorylation of Ser214 plays a
hyperphosphorylation of tau in AD brain are significant role in regulating the association of
equivocal. For example, immunoblot analyses of tau with microtubules (64). Using 32P-labelled
supernatant and particulate fractions from AD cells and phosphopeptide mapping, it was
and control brains with antibodies to GSK-3β demonstrated that during mitosis there is a
revealed a 30% decrease in GSK-3β immuno- significant increase in the phosphorylation of
reactivity in AD as compared to control brain. Ser214, which is the site on tau which is pre-
Additionally there was a negative correlation ferentially phosphorylated by protein kinase A
between GSK-3β levels and numbers of NFTs (135). Phosphorylation of tau by protein kinase
(9). In contrast, Pei et al. (118) found a 50% A in vitro reduces its ability to nucleate
increase in GSK-3β levels in the post- microtubules and promote microtubule assembly
synaptosomal supernatant from AD brains as (16). Because tau is an in situ substrate of
compared to control brain using an indirect protein kinase A (35,91), it is likely that protein
ELISA. However, no increase in GSK-3β kinase A is the protein kinase which phos-
enzyme activity in AD as compared to control phorylates tau on Ser214 in these mitotic LAN-5
brains was observed (118). Considering these and tau-transfected CHO cells (64), but further
disparate findings, it is clear that more research is validation is required.
required to determine whether GSK-3β It has also been suggested that increased
contributes to tau hyperphosphorylation in AD. oxidative stress in AD brain may contribute to
A family of novel protein kinases named the hyperphosphorylation of tau. However, the
MARK (MAP/microtubule affinity-regulating majority of studies indicate that it is unlikely that
kinase) have been implicated in phosphorylating acute oxidative stress results in increased tau
the critical Ser262 residue of tau (30). This phosphorylation. For example, both in rat brain
residue is located in the KXGS sequence of the primary neuronal cultures (27) and in cultured
first repeat that is part of the microtubule-binding human neuroblastoma MSN cells (82), acute
domain of tau. Phosphorylation at this site is oxidative stress resulted in tau dephos-
elevated in AD (107) and Ser262 phosphorylation phorylation. However, enhanced tau phosphory-
significantly decreases tau-microtubule binding lation and/or aggregation in response to low
(11,160). Transient expression of tau in CHO level, chronic oxidative stress cannot be ruled
cells that were stably transfected with the MARK out.
protein resulted in a disruption of the microtubule Sadot et al. (128) provided evidence for a link
array, suggesting that MARK phosphorylated tau between the cholinergic signal transduction
and decreased its ability to bind and stabilize system and the neuronal cytoskeleton. The
microtubules in situ (30). Interestingly, MARK authors transfected rat PC12 cells with the m1
was also found to be inactivated by protein muscarinic acetylcholine receptor. They then sti-
phosphatase 2A (PP2A) (30), which is sig- mulated the cells with two acetylcholine agonists,
nificantly decreased in AD brain (45,46,147). carbachol, and AF102B, which selectively
Therefore, the decrease in PP2A activity in AD activates m1 receptors, and examined the
brain could contribute to the hyperphos- phosphorylation state of tau. Tau phos-
336 G.V.W. Johnson and J.A. Hartigan / Tau Protein in Normal and Alzheimer’s Disease Brain

phorylation decreased at the Tau-1 and AT8 sites, phorylation, but it completely inhibited calpain-
which include the Ser202 residue (128). This is mediated tau proteolysis. These findings indicate
the first study to provide a link between the that dephosphorylation of tau by PP2B at specific
cholinergic signal transduction pathway and tau sites is necessary for calpain-mediated de-
phosphorylation, and suggests that decreased gradation of tau (173).
cholinergic activity could contribute to an In summary, there is a complex interplay
increase in tau phosphorylation. between the protein phosphatases and kinases in
Hall et al. (50) described a useful model in the brain that modulate tau phosphorylation, and
which to study the cellular mechanisms under- it is likely that no one particular enzyme is
lying tau hyperphosphorylation. They injected responsible for the hyperphosphorylated state of
plasmids encoding human tau protein into tau in AD. The in situ studies cited here,
lamprey central neurons in vivo. In this model demonstrate the complexity of the systems in
the human tau was phosphorylated at the PHF-1 living cells. Needless to say, there is much work
(Ser396/404) and Tau-1/AT8 (Ser199/202) sites. left to be done to understand the role of kinases
Accumulations of tau were also observed and the and phosphatases in modulating the phosphory-
cells exhibited degenerative changes (50). This lation state of tau in situ.
model may be useful in studying tau
phosphorylation in situ and the relationship
between increased tau phosphorylation and PHOSPHORYLATION OF PHF-tau
neurodegeneration.
In addition to protein kinases, protein Recently, a number of new monoclonal tau
phosphatases play an important role in antibodies that recognize specific phospho-
modulating tau phosphorylation. Co-expression epitopes or conformations have been described.
of tau and SV40 small t antigen, a PP2A Antibodies directed against specific phospho-
inhibitor, in non-neuronal cells resulted in an epitopes in tau have proven to be excellent tools
increase in tau phosphorylation at a number of in the elucidation of tau phosphorylation under
sites, including Ser199, Ser202, Thr205, Ser396 and physiological and pathological conditions. A
Ser404. This phosphorylation also resulted in a summary of some of these numerous phospho-
loss of microtubule stability and the dissociation specific tau antibodies is shown in Table 2. PHF-
of tau from the microtubules (147). In NT2N and 9 specifically recognizes phosphorylated Ser404
SH-SY5Y cells, depolymerization of micro- (175), while AP422 recognizes phosphorylated
tubules with nocadozole or stabilization of Ser422 (54). AD2 is another phosphorylation
microtubules with taxol results in the dependent monoclonal antibody that recognizes
dephosphorylation of tau at several Ser/Thr-Pro phosphorylated Ser396 and Ser404 (18). Hoffmann
epitopes by PP2A (103,174). Because a small et al. (58) describe two antibodies that are unique
subpopulation of the total PP2A associates with in that they are dependent on the phosphorylation
microtubules (146), it can be suggested that the of two residues. AT10 (also referred to as
polymerization state of microtubules modulates AT100 in (176)) requires the simultaneous
the phosphorylation state of tau indirectly phosphorylation of Thr212 and Ser214, while PHF-
through its effects on PP2A. The calcium- 27 requires the phosphorylation of the primary
calmodulin dependent phosphatase calcineurin, site Thr231 and the subsite Ser235 (58). In
or PP2B, also regulates the phosphorylation state addition, both AT10 (AT100) and PHF-27 only
of tau. Treatment of PC12 cells with maitotoxin recognized PHF-tau and not fetal or biopsy
increases intracellular calcium levels and resulted derived tau (58,176). These data indicate that
in tau dephosphorylation and calpain-mediated concurrent phosphorylation of tau on these sites
tau degradation. Treatment of the cells with a occurs in pathological conditions such as AD.
calcineurin inhibitor, FK502, prior to maitotoxin MC-1, a conformation dependent monoclonal
resulted in a partial inhibition of tau dephos- antibody, recognizes a tau conformation similar
G.V.W. Johnson and J.A. Hartigan / Tau Protein in Normal and Alzheimer’s Disease Brain 337

Table 2
Phosphoepitopes Recognized by Different Tau Antibodies. Indicates a phosphorylation pair, where
phosphorylation of both sites is required for optimal immunoreactivity. Indicates dephosphorylation. Stars
represent phosphorylation, with smaller stars indicating a subsequent minor phosphorylation site. The references
cited in the table are as follows: a, (154); b, (18); c, (54); d, (64); e, (176); f, (40); g, (55); h, (116); i, (100); j, (175);
k, (58); l, (143); m, (59); n, (141)

to that recognized by Alz-50 (71). As was ment and remain phosphorylated throughout the
mentioned earlier, Alz-50 recognizes two NFT stages. In addition, Thr231 and Ser396 are
discontinuous segments of the tau molecule phosphorylated only in the more advanced stages
(21). TG-3 is also a conformation dependent of NFTs (80). There is also evidence from AD
monoclonal antibody that recognizes a tau brain that casein kinase I selectively associates
conformation found specifically in PHF-tau (72). with PHFs and may contribute to tau hyper-
A subpopulation of vulnerable hippocampal phosphorylation (85).
CA1 pyramidal neurons from AD brain stained One of the intriguing aspects of PHF-tau is
positively with PS 413, an antibody that that it shares many of the same phosphorylated
recognizes tau when Ser413 is phosphorylated sites as fetal tau. Fetal tau is also phosphorylated
(143). Kimura et al. (80) investigated the to a significantly greater extent than control
changes in tau phosphorylation as the NFTs human tau (77,169), which has led some to
develop. Phosphorylation of sites Ser199, Ser202, hypothesize that a recapitulation of a fetal-like
Ser409, and Ser422 occurs early in tangle develop- state occurs in AD which is characterized by the
338 G.V.W. Johnson and J.A. Hartigan / Tau Protein in Normal and Alzheimer’s Disease Brain

inappropriate expression of mitotic proteins. The in tau being modified in such a way that it could
antibody MPM2 recognizes phosphoepitopes that not be efficiently dephosphorylated (98). In P19
are found predominantly in mitotic cells (163). neuroglial cultures, HNE was found to stimulate
Immunohistochemical analyses of AD hippo- the formation of ubiquitinated cross-linked
campus revealed that both MPM2 and AT8, proteins, including tau, but was also a potent
which recognizes tau when Ser202/Thr205 are cytotoxin (105). Although these studies are
phosphorylated, stained NFTs (83,166) and co- intriguing, future work is necessary to determine
localized to pyramidal neurons (83). MPM2 also if lipid peroxidation or HNE-linked modifications
reacts with PHF-tau, but not normal fetal or adult contribute to the tau pathology in AD brain.
rat tau (83). It has also been shown that the Glycation of tau is another potential
presence of M-phase cdc2/cyclin B1 kinase modification that may be involved in the
complex is elevated in AD brain compared to formation of PHFs. As reported previously, tau
controls, and the proteins were enriched in is a good candidate for glycation, and PHF-tau
neurons with NFTs and in neurons susceptible to was found to be glycated, while normal tau was
NFTs (165). Additionally, it has been established not (74). Glycation is the non-enzymatic ad-
that certain cell cycle proteins such as cyclin D, dition of a reducing sugar to a protein, often on a
cdk4, and proliferating cell nuclear antigen are lysine residue, and is a modification that often
aberrantly expressed in AD brain (20). These occurs in long-lived proteins during the aging
finding suggest that the inappropriate expression process (10). Nacharaju et al. (112) characterized
of mitotic proteins may contribute to the the sites on tau that are glycated in vitro to
hyperphosphorylation of tau and the neuro- determine if the repeat regions are involved.
degenerative process in AD. Using the longest isoform of tau, it was
It has been well-established that PHF-tau demonstrated that six sites within the repeat
exhibits significantly reduced electrophoretic regions (Lys259, 280, 281, 347, 353, and 369) were gly-
mobility and often presents as a triplet, with cated (112). Since this area of the tau molecule is
relative molecular masses from 60 to 70 kDa important for binding and stabilizing micro-
(88). Recently, the presence of an additional tubules, glycation here could disrupt normal tau
∼ 74 kDa hyperphosphorylated tau form was function and contribute to the formation of PHFs
detected in the youngest and most severely and NFTs. However, because it is long-lived,
affected AD cases, and apparently corresponded lysine-rich proteins that are modified, it is
to the longest tau isoform found in brain. conceivable that glycation of PHF-tau occurs
Furthermore, based on isoelectric points, it was subsequent to PHF and NFT formation. There is
determined that PHF-tau from older AD patients also preliminary data suggesting that the levels of
was less phosphorylated than PHF-tau from oxidized products from non-enzymatic glycation
younger AD cases (140). These findings indicate are not elevated in AD brain compared to
age of onset and duration of the disease are controls (136). Therefore, the modification of
important factors to consider when evaluating the proteins by glycation in AD brain may be a
characteristics of PHF-tau. selective process and not the result of global
changes.
Tau is apparently modified by glycosylation.
OTHER TAU MODIFICATIONS Ser(Thr)-O-linked N-acetylglycosamination is a
dynamic and abundant posttranslational modifi-
Lipid peroxidation, and the formation of its cation that can be reciprocal with Ser/Thr phos-
aldehydic by-product 4-hydroxynonenal (HNE), phorylation. In an initial study, bovine tau was
occur in response to oxidative stress and have found to be modified by O-linked glycosylation
been implicated in the modifications of tau in AD at multiple sites, and it is likely that this
brain. In a preliminary study, treatment of modification may play a role in modulating tau
cultured rat hippocampal cells with HNE resulted function (7). The modification of PHF-tau, but
G.V.W. Johnson and J.A. Hartigan / Tau Protein in Normal and Alzheimer’s Disease Brain 339

not normal tau, by N-linked glycosylation has phorylation of tau in response to Aβ treatment
also been reported (168). In contrast to O-linked resulted in a loss of microtubule binding capacity
glycosylation, N-linked glycosylation usually and accumulation of tau in the somatodendritic
occurs co-translationally and is a considerably compartment (19). Similarly, treatment of the
less dynamic modification. Interestingly, degly- hybrid septal cell line, SN56, with Aβ1–40 resulted
cosylation of PHFs converted them into straight in tau phosphorylation at Ser199/202 and Ser396, and
filaments and therefore it was suggested that N- cell toxicity. However, treatment with Aβ1–28,
linked glycosylation may contribute to the which is non-amyloidogenic, had no significant
maintenance of PHF-structure (168). effects on tau phosphorylation (87). Interesting-
ly, it was recently reported that neuronal cell
cultures were protected from Aβ toxicity by
Aβ, CALCIUM HOMEOSTASIS AND taxol, a microtubule-stabilizing drug (104).
TAU HYPERPHOSPHORYLATION There is also some data to indicate that increased
Aβ deposition in animal models also results in
Aβ constitutes the core of the extracellular increased tau phosphorylation. For example, a
neuritic plaques, another neuropathic hallmark of recent study in transgenic mice overexpressing
AD in addition to the NFTs. Aβ is a 39-43 amino human APP containing either the Swedish double
acid peptide that results from the cleavage of the mutation at positions 670/671 alone or in
695-770 amino acid amyloid precursor protein combination with the London V717I mutation,
(APP) (for a review see (52,96,137). Because demonstrated that the dystrophic neurites that
neuritic plaques and NFTs occur in AD brain, the developed around the plaques stained positive for
interrelationship between the two major specific phosphorylated forms of tau. These
components of these structures, tau and Aβ, is immunohistochemical findings were corraborated
being examined. Diffuse and dense neuritic by immunoblot analysis which also revealed an
plaques in AD brain immunohistochemically increase in tau phosphorylation (152).
label with antibodies against PHF-tau (131) and One possible mechanism by which Aβ may
PHF-tau positive neurites are present in and induce increases in tau phosphorylation is by
around all subtypes of plaques (153). Converse- perturbing calcium homeostasis which would
ly, antibodies to an epitope between residues 713 result in inappropriate increases in the activities
and 723 of APP770 consistently labeled PHFs in of certain protein kinases contributing to tau
the AD brain (38). In vitro, a peptide segment hyperphosphorylation. Further, it has been
corresponding to residues 713 to 730 of APP770 hypothesized that Aβ-induced perturbation of
(which is in the C-terminus region) bound to tau calcium homeostasis could make the cells more
and formed dense fibrillary assemblies containing vulnerable to other potentially “toxic” stimuli,
both tau and APP (38). Domains of APP that are including glutamate. Treatment of human cere-
C-terminal of the Aβ-containing segment bind to bral cortical or rat hippocampal cell cultures with
tau and tubulin in vitro (67), and the non- Aβ resulted in elevated intracellular calcium
amyloidogenic Aβ1–28 also bound directly to levels over those of resting cells. Addition of Aβ
tubulin (67). These data indicate that direct also made the cells more vulnerable to
interactions between tau and specific segments of neurotoxicity induced by glutamate, kainate or N-
APP, including the Aβ domain, may occur both methyl-D-aspartate (NMDA) (97,99). Interes-
normally and/or pathologically in AD brain. tingly, rat hippocampal cells were protected
There have also been numerous studies against Aβ induced calcium perturbation by basic
examining the possible effects of Aβ on tau fibroblast growth factor (bFGF), and therefore it
phosphorylation. Treatment of fetal rat hippo- was suggested that bFGF may be involved in
campal and human cortical neurons with fibrillar stabilizing intracellular calcium levels (99).
Aβ resulted in an increase of tau phosphorylation Although treatment with Aβ may result in
at Ser202/Thr205 and Ser396/Ser404 (19). The phos- increases in intracellular calcium, this may not be
340 G.V.W. Johnson and J.A. Hartigan / Tau Protein in Normal and Alzheimer’s Disease Brain

the mechanism by which it induces the putative TAU IN OTHER


increases in tau phosphorylation. For example, NEURODEGENERATIVE DISEASES
treatment of human neuroblastoma cells with
either a calcium ionophore or Aβ25–35 resulted in Hyperphosphorylated forms of tau occur in
an increase in intracellular calcium levels and many neurodegenerative diseases besides AD,
increased tau phosphorylation at specific epitopes and in almost all cases accumulates as
(142). The increase in tau phosphorylation elicited filamentous lesions. The presentation of the
by the calcium ionophore was blocked by the hyperphosphorylated tau shows some variability
calcium chelator EGTA, however the Aβ25–35 between the diseases, both in terms of
induced increase in tau phosphorylation was not, electrophoretic mobility (i.e., migrating as two,
suggesting that it was not mediated by calcium three, or four bands) and in the ultrastructural
(142). Further, there is some evidence to suggest characteristics of the filaments (e.g., straight
that Aβ does not increase intracellular calcium versus paired helical, filament dimensions, etc.)
concentrations. For example, Aβ25–35 treatment of (32,84,101,132). However, it has been hypo-
primary rat cortical neurons (27) and Aβ1–42 thesized that because of the clinical and
treatments of human neuron-like NT2N cells (37) pathological overlap among neurodegenerative
did not result in an increase in intracellular disorders with extensive tau pathology, such as
calcium concentrations. The conflicting nature of AD, corticobasal degeneration (CBD), progres-
these findings clearly indicates the need for sive supernuclear palsy (PSP), and Guam amyo-
additional studies to clarify the involvement of trophic lateral sclerosis/Parkinsonism-dementia
calcium-mediated events in Aβ-induced increases complex (ALS/PDC), that it is likely that they
in tau phosphorylation. share some etiological correlates (32). Interes-
Another possible mechanism by which Aβ may tingly, there is growing evidence that mutations
induce increases in tau phosphorylation is by in the tau gene may be a contributing factor in
activation of GSK-3β. GSK-3β is one of the few several neurodegenerative diseases with ex-
protein kinases shown to phosphorylate tau in situ tensive tau pathology. For example, intronic
(17,51,59,60,161,167). Exposure of rat hippo- polymorphisms in the tau gene may be involved
campal cell culture to Aβ resulted in GSK-3β in PSP (23,57).
activation and increased tau phosphorylation Although tau mutations have been implicated
(61). In addition, activation of GSK-3β by Aβ in certain familiar forms of PSP (23,57), strong
may contribute to neuronal death. Treatment of evidence for a role of tau mutations in certain
rat hippocampal cells with GSK-3β antisense families with autosomal dominant forms of
oligonucleotides prevented Aβ-induced neuronal presenile frontotemporal dementia (FTD) has
death (156). Furthermore, it was demonstrated recently been described. Several pedigrees of
that exposure of rat hippocampal cells to Aβ25–35 FTD have been linked to chromosome 17q21
results in the inactivation of phosphatidylinositol-3 (12,56,111,149) which is also the location of the
(PI-3) kinase (155). PI-3 kinase activates Akt tau gene (1). This subset of frontotemporal
which phosphorylates GSK-3β on Ser9 leading to dementias is collectively referred to as “fronto-
inactivation and decreased tau phosphorylation temporal dementia and Parkinsonism linked to
(60). Therefore, inactivation of PI-3 kinase by Aβ chromosome 17” (FTDP-17). Several of these
would result in an increase of GSK-3β activity and families have abundant widespread tau-positive
subsequent increase in tau phos-phorylation. fibrillary deposits in both neurons and glia
These findings have lead to the hypothesis that Aβ (12,111,124,149). Although abundant neuro-
decreases PI-3 kinase activity thus disinhibiting fibrillary inclusions are present in these FTDs,
GSK-3β which in turn results in increased tau amyloid plaques were virtually absent. In recent
phosphorylation, impaired axonal transport and reports, specific mutations in the tau gene have
ultimately somatodendritic tau accumulation and been shown to occur in a number of these
neuronal death (61, 155,156). families with FTDP-17 (122,150). Mutations
G.V.W. Johnson and J.A. Hartigan / Tau Protein in Normal and Alzheimer’s Disease Brain 341

have been found in the tau introns (62,150), autopsy that AD can be diagnosed conclusively.
including the 5' splice site of exon 10 (62), as Therefore, the presence of biomarkers specific
well as in the coding region of the tau gene (122). for AD have been sought. In 1993, Vander-
It was hypothesized that mutations of the tau meeren et al. (162) developed a sensitive sand-
introns may be responsible for the increase in wich enzyme-linked immunosorbent assay to
synthesis of the larger isoforms of tau and result detect the low levels of tau found in the CSF, and
in abnormal tau aggregation (62,150). Several with this assay found an increase in CSF tau
mutations occurred within the microtubule- levels in AD patients (162). Shortly thereafter,
binding domains of tau, and therefore could Mandelkow and Mandelkow (94) suggested that
disrupt tau-microtubules interactions and thus tau levels in the CSF may be used as a biomarker
contribute to tau aggregation and neuro- for AD, when they also found an increase of tau
degeneration (62,150). in the CSF of AD patients (94). Further, the tau
It is intriguing that in these FTDP-17 diseases, found in the CSF of both controls and AD
as well as a number of other neurodegenerative patients is not full length tau, but a proteolytic
diseases including CBD, PSP and ALS/PDC, product containing the N-terminal end of the
there is extensively neurofibrillary pathology molecule (73). There are now numerous articles
without abundant amyloid plaques (148). These on tau in the CSF, and the overwhelming
data suggest that in AD the amyloid plaques and consensus is that tau levels in the CSF of AD
NFTs are independent lesions and one is not the patients are significantly increased over the CSF
sole “cause” of the other. This is not to rule out tau levels of normal controls (e.g. 36,70,106,108,
the possibility that the consequences of the 110,127,158,164). CSF tau levels from AD
presence of one lesion may potentiate the patients were also elevated over those with multi-
development of the other. For example, Aβ infarct dementia (158), cerebrovascular disease
could, through some mechanism, contribute to dementia (106), non-AD dementia (164), and
the increase in tau phosphorylation (e.g. see mild cognitive impairment and other dementing
preceding section). Indeed, in one of the amyloid diseases (15). Galasko et al. (36) found an
precursor protein transgenic mice models, increase in CSF tau levels from mildly impaired
increased tau phosphorylation was observed, patients with AD, suggesting that elevated levels
especially around the amyloid plaques (152), of CSF tau occur very early in the disease.
suggesting that Aβ deposition may facilitate the However, one study reported that CSF tau levels
hyperphosphorylation of tau. Therefore, further from AD patients were as high as those from
study of the interrelationship between Aβ and tau patients with dementia with Lewy bodies
pathologies is essential. (5/6). Elevated levels of tau in CSF were also
found in patients suffering from Creutzfeldt-
Jakob Disease (CJD) (115). Blennow et al. (14)
TAU IN THE CEREBROSPINAL FLUID found considerable overlap in CSF tau levels
with AD patients and those with other forms of
The primary mechanism by which an dementia including vascular dementia and frontal
antemortem diagnosis of AD is made is through lobe dementia (14). These studies indicate that
the use of neuropsychological analysis. Although elevated CSF tau levels alone cannot be used to
neuropsychological testing is extremely useful, conclusively diagnose AD.
definitive diagnosis of AD based on objective In addition to increases in CSF tau, decreases
biological markers would greatly facilitate the in Aβ1–42 peptide in the CSF may also provide
early identification of AD patients. The ability to support for a diagnosis of AD. CSF levels of
accurately identify AD patients early in the soluble APP have been found to be decreased in
course of the disease will become even more AD patients as compared to controls (86,121).
imperative as effective treatments of the disease However, a more consistent and reliable
become available. Presently, it is not until difference has been found in CSF Aβ1–42
342 G.V.W. Johnson and J.A. Hartigan / Tau Protein in Normal and Alzheimer’s Disease Brain

levels. The CSF levels of Aβ1–42 have been CONCLUSIONS AND SPECULATIONS
found to be significantly decreased in AD
patients compared to depressed control subjects It is becoming increasingly evident that
(133) and normal controls (108,157). The classification of tau simply as a microtubule
results on the Aβ1–40 segment are mixed, with binding and stabilizing protein is insufficient.
some showing no change in AD as compared to Recent work strongly indicates that tau is
controls (157) and others showing decreased multifunctional and likely to play a significant
CSF Aβ1–40 in AD over depressed controls role in a variety of neuronal functions. In order
(133). These data indicate that increased CSF to identify the mechanisms responsible for the
tau levels in combination with decreased CSF abnormal accumulation of tau in AD and other
Aβ1–42 levels may be useful indicators in the neurodegenerative diseases, a clearer under-
diagnosis of AD. standing of tau’s functions, and the modifications
Apolipoprotein E alleles have also been which regulate these functions, is required.
detected in the CSF. A full discussion of the Tau is a highly phosphorylated protein, and it
role of apolipoprotein E is beyond the scope of is readily evident that site-specific phos-
this paper. Suffice it to say that the presence of phorylation of the molecule is a very dynamic
the apolipoprotein E-4 allele (apoE4) is a process. Recent studies have begun to provide
genetic risk factor for sporadic and familiar late- significant insight into the protein kinases and
onset AD (24,130). Numerous researchers have phosphatases that modulate the phosphorylation
measured the levels of both tau and apoE4 in the state of tau in situ. However, much remains to be
CSF to determine whether there is a connection learned about the dynamic interplay between
between the two. To date, the results of these specific protein kinases and phosphatases in
experiments are mixed, with some authors normal and pathological conditions, as well as
finding that increased CSF tau levels are the identity of these enzymes which modulate the
correlated with increased CSF apoE4 levels in vivo phosphorylation state of tau. It is also
(15,44,90) and others finding no relationship evident that phosphorylation is not the only
between CSF tau levels and CSF apoE4 levels regulatory posttranslational modification of tau.
(6,108,114). Glycosylation, as well as other modifications, are
Another area where the results are mixed is in likely to regulate tau function and localization.
whether the CSF tau levels correlate with the Elucidating the posttranslational processing of
severity of cognitive impairment or dementia. tau is critically important to our understanding of
Tato et al. (158) found that CSF tau levels the involvement of tau in the neuropathology of
correlated positively with the evolution of the AD.
disease and negatively with a mental state The recent findings that mutations in the tau
examination. However, others have found no gene have been linked to several autosomal
correlation between CSF tau levels and degree dominant FTDP-17’s is particularly exciting. In
of cognitive impairment or severity of dementia at least two of the pedigrees, fibrillary deposits
(110,114,127). composed of hyperphosphorylated tau are
At this point, it is clear that elevated CSF tau prevalent but there is a marked absence of Aβ
levels are insufficient to make a definitive deposits. These findings indicate that abnor-
diagnosis of AD. The combination of elevated malities in the processing and function of tau are
CSF tau levels and decreased CSF Aβ1–42 levels likely to contribute to the neurodegenerative
increases the reliability of an AD diagnosis but process. The studies strongly suggest that in AD
is still not sufficiently definitive to make a and these other neurodegenerative diseases, the
conclusive diagnosis. The ability to use CSF tau abnormal modification of tau and eventual
levels in conjunction with other markers may aggregation into neurofibrillary lesions is not just
increase the accuracy of a diagnosis of AD, an “outcome” of the neuronal death process, but
however more studies are clearly necessary. rather plays a critical important role in the
G.V.W. Johnson and J.A. Hartigan / Tau Protein in Normal and Alzheimer’s Disease Brain 343

neurodegenerative process. However, before any Role of glycosaminoglycans in determining the


definitive conclusions can be reached, it is clear helicity of paired helical filaments, Am J Pathol
that further studies are required. 151 (1997) 1115–1122.
9. Baum L, Hansen L, Masliah E, Saitoh T,
Glycogen synthase kinase 3 alteration in
Alzheimer disease is related to neurofibrillary
ACKNOWLEDGMENTS tangle formation, Mol Chem Neuropathol 29
(1996) 253–261.
Our research was supported by grants from the 10. Baynes JW, Role of oxidative stress in
NIH (AG12396, AG06569, NS27538 and development of complications in diabetes,
NS35060). Diabetes 40 (1991) 405–412.
11. Biernat J, Gustke N, Drewes G, Mandelkow EM,
Mandelkow E, Phosphorylation of Ser262
strongly reduces binding of tau to microtubules:
REFERENCES distinction between PHF-like immunoreactivity
and microtubule binding, Neuron 11 (1993) 153–
1. Abel KJ, Boehnke M, Prahalad M, Ho P, Flejter 163.
WL, Watkins M, VanderStoep J, 12. Bird TD, Wijsman EM, Nochlin D, Leehey M,
Chandrasekharappa SC, Collins FS, Glover TW, Sumi SM, Payami H, Poorkaj P, Nemens E,
et al. A radiation hybrid map of the BRCA1 Rafkind M, Schellenberg GD, Chromosome 17
region of chromosome 17q12-q21, Genomics 17 and hereditary dementia: linkage studies in three
(1993) 632–641. non-Alzheimer families and kindreds with late-
2. Alonso AC, Grundke-Iqbal I, Iqbal K, onset FAD, Neurology 48 (1997) 949–954.
Alzheimer’s disease hyperphosphorylated tau 13. Black MM, Slaughter T, Moshiach S, Obrocka
sequesters normal tau into tangles of filaments M, Fischer I, Tau is enriched on dynamic
and disassembles microtubules, Nat Med 2 microtubules in the distal region of growing
(1996) 783–787. axons, J Neurosci 16 (1996) 3601–3619.
3. Andreadis A, Wagner BK, Broderick JA, Kosik 14. Blennow K, Wallin A, Agren H, Spenger C,
KS, A tau promoter region without neuronal Siegfried J, Vanmechelen E, Tau protein in
specificity, J Neurochem 66 (1996) 2257–2263. cerebrospinal fluid: a biochemical marker for
4. Appelt DM, Balin BJ, The association of tissue axonal degeneration in Alzheimer disease? Mol
transglutaminase with human recombinant tau Chem Neuropathol 26 (1995) 231–245.
results in the formation of insoluble filamentous 15. Blomberg M, Jensen M, Basun H, Lannfelt L,
structures, Brain Res 745 (1997) 21–31. Wahlund LO, Increasing cerebrospinal fluid tau
5. Arai H, Morikawa Y, Higuchi M, Matsui T, levels in a subgroup of Alzheimer patients with
Clark CM, Miura M, Machida N, Lee VM, apolipoprotein E allele epsilon 4 during 14
Trojanowski JQ, Sasaki H, Cerebrospinal fluid months follow-up, Neurosci Lett 214 (1996)
tau levels in neurodegenerative diseases with 163–166.
distinct tau-related pathology, Biochem Biophys 16. Brandt R, Lee G, Teplow DB, Shalloway D,
Res Commun 236 (1997) 262–264. Abdel-Ghany M, Differential effect of
6. Arai H, Terajima M, Miura M, Higuchi S, phosphorylation and substrate modulation on
Muramatsu T, Matsushita S, Machida N, tau’s ability to promote microtubule growth and
Nakagawa T, Lee VM, Trojanowski JQ, Sasaki nucleation, J Biol Chem 269 (1994) 11776–
H, Effect of genetic risk factors and disease 11782.
progression on the cerebrospinal fluid tau levels 17. Brownlees J, Irving NG, Brion JP, Gibb BJ,
in Alzheimer’s disease, J Am Geriatr Soc 45 Wagner U, Woodgett J, Miller CC, Tau
(1997) 1228–1231. phosphorylation in transgenic mice expressing
7. Arnold CS, Johnson GV, Cole RN, Dong DLY, glycogen synthase kinase-3beta transgenes,
Lee M, Hart GW, The microtubule-associated Neuroreport 8 (1997) 3251–3255.
protein tau is extensively modified with O-linked 18. Buee-Scherrer V, Condamines O, Mourton-Gilles
N-acetylglucosamine, J Biol Chem 271 (1996) C, Jakes R, Goedert M, Pau B, Delacourte A,
28741–28744. AD2, a phosphorylation-dependent monoclonal
8. Arrasate M, Perez M, Valpuesta JM, Avila J, antibody directed against tau proteins found in
344 G.V.W. Johnson and J.A. Hartigan / Tau Protein in Normal and Alzheimer’s Disease Brain

Alzheimer’s disease, Brain Res Mol Brain Res 39 Microfilament-associated growth cone
(1996) 79–88. component depends upon Tau for its intracellular
19. Busciglio J, Lorenzo A, Yeh J, Yankner BA, localization, Cell Motil Cytoskeleton 29 (1994)
beta-amyloid fibrils induce tau phosphorylation 117–130.
and loss of microtubule binding, Neuron 14 30. Drewes G, Ebneth A, Preuss U, Mandelkow EM,
(1995) 879–888. Mandelkow E, MARK, a novel family of protein
20. Busser J, Geldmacher DS, Herrup K, Ectopic cell kinases that phosphorylate microtubule-
cycle proteins predict the sites of neuronal cell associated proteins and trigger microtubule
death in Alzheimer’s disease brain, J Neurosci 18 disruption, Cell 89 (1997) 297–308.
(1998) 2801–2807. 31. Dudek SM, Johnson GV, Transglutaminase
21. Carmel G, Mager EM, Binder LI, Kuret J, The catalyzes the formation of sodium dodecyl
structural basis of monoclonal antibody Alz50’s sulfate-insoluble, Alz-50-reactive polymers of
selectivity for Alzheimer’s disease pathology, J tau, J Neurochem 61 (1993) 1159–1162.
Biol Chem 271 (1996) 32789–32795. 32. Feany MB, Dickson DW, Neurodegenerative
22. Chen WT, Liu WK, Yen SH, Expression of tau disorders with extensive tau pathology: a
exon 8 in different species, Neurosci Lett 172 comparative study and review, Ann Neurol 40
(1994) 167–170. (1996) 139–148.
23. Conrad C, Andreadis A, Trojanowski JQ, 33. Felgner H, Frank R, Biernat J, Mandelkow EM,
Dickson DW, Kang D, Chen X, Wiederholt W, Mandelkow E, Ludin B, Matus A, Schliwa M,
Hansen L, Masliah E, Thal LJ, Katzman R, Xia Domains of neuronal microtubule-associated
Y, Saitoh T, Genetic evidence for the proteins and flexural rigidity of microtubules, J
involvement of tau in progressive supranuclear Cell Biol 138 (1997) 1067–1075.
palsy, Ann Neurol 41 (1997) 277–281. 34. Flanagan LA, Cunningham CC, Chen J,
24. Corder EH, Saunders AM, Strittmatter WJ, Prestwich GD, Kosik KS, Janmey PA, The
Schmechel DE, Gaskell PC, Small GW, Roses structure of divalent cation-induced aggregates of
AD, Haines JL, Pericak-Vance MA, Gene dose PIP2 and their alteration by gelsolin and tau,
of apolipoprotein E type 4 allele and the risk of Biophys J 73 (1997) 1440–1447.
Alzheimer’s disease in late onset families [see 35. Fleming LM, Johnson GV, Modulation of the
comments], Science 261 (1993) 921–923. phosphorylation state of tau in situ: the roles of
25. Cross DA, Alessi DR, Cohen P, Andjelkovich M, calcium and cyclic AMP, Biochem J 309 (1995)
Hemmings BA, Inhibition of glycogen synthase 41–47.
kinase-3 by insulin mediated by protein kinase B, 36. Galasko D, Clark C, Chang L, Miller B, Green
Nature 378 (1995) 785–789. RC, Motter R, Seubert P, Assessment of CSF
26. Cunningham CC, Leclerc N, Flanagan LA, Lu M, levels of tau protein in mildly demented patients
Janmey PA, Kosik KS, Microtubule-associated with Alzheimer’s disease [see comments],
protein 2c reorganizes both microtubules and Neurology 48 (1997) 632–635.
microfilaments into distinct cytological structures 37. Gao Z-Y, Collins HW, Matschinsky FM, Lee
in an actin-binding protein-280-deficient VM, Wolf BA, Cytotoxic effect of b-amyloid on
melanoma cell line, J Cell Biol 136 (1997) 845– human differentiated neuron is not mediated by
857. cytoplasmic Ca2+ accumulation, J Neurochem 70
27. Davis DR, Anderton BH, Brion JP, Reynolds (1998) 1394–1400.
CH, Hanger DP, Oxidative stress induces 38. Giaccone G, Pedrotti B, Migheli A, Verga L,
dephosphorylation of tau in rat brain primary Perez J, Racagni G, Smith MA, Perry G, De
neuronal cultures, J Neurochem 68 (1997) 1590– Gioia L, Selvaggini C, Salmona M, Ghiso J,
1597. Frangione B, Islam K, Bugiani O, Tagliavini F,
28. Davis DR, Brion JP, Couck AM, Gallo JM, beta PP and Tau interaction, A possible link
Hanger DP, Ladhani K, Lewis C, Miller CC, between amyloid and neurofibrillary tangles in
Rupniak T, Smith C, et al. The phosphorylation Alzheimer’s disease, Am J Pathol 148 (1996) 79–
state of the microtubule-associated protein tau as 87.
affected by glutamate, colchicine and beta- 39. Goedert M, Hasegawa M, Jakes R, Lawler S,
amyloid in primary rat cortical neuronal cultures, Cuenda A, Cohen P, Phosphorylation of
Biochem J 309 (1995) 941–949. microtubule-associated protein tau by stress-
29. DiTella M, Feiguin F, Morfini G, Caceres A, activated protein kinases, FEBS Lett 409 (1997)
G.V.W. Johnson and J.A. Hartigan / Tau Protein in Normal and Alzheimer’s Disease Brain 345

57–62. when overexpressed in lamprey central neurons


40. Goedert M, Jakes R, Crowther RA, Cohen P, in situ, Proc Natl Acad Sci USA 94 (1997) 4733–
Vanmechelen E, Vandermeeren M, Cras P, 4738.
Epitope mapping of monoclonal antibodies to the 51. Hanger DP, Hughes K, Woodgett JR, Brion JP,
paired helical filaments of Alzheimer’s disease: Anderton BH, Glycogen synthase kinase-3
identification of phosphorylation sites in tau induces Alzheimer’s disease-like phosphorylation
protein, Biochem J 301 (1994) 871–877. of tau: generation of paired helical filament
41. Goedert M, Jakes R, Spillantini MG, Hasegawa epitopes and neuronal localisation of the kinase,
M, Smith MJ, Crowther RA, Assembly of Neurosci Lett 147 (1992) 58–62.
microtubule-associated protein tau into 52. Hardy J, Amyloid, the presenilins and
Alzheimer-like filaments induced by sulphated Alzheimer’s disease, Trends Neurosci 20 (1997)
glycosaminoglycans [see comments], Nature 383 154–159.
(1996) 550–553. 53. Hasegawa M, Crowther RA, Jakes R, Goedert M,
42. Goedert M, Spillantini MG, Jakes R, Rutherford Alzheimer-like changes in microtubule-
D, Crowther RA, Multiple isoforms of human associated protein Tau induced by sulfated
microtubule-associated protein tau: sequences glycosaminoglycans, Inhibition of microtubule
and localization in neurofibrillary tangles of binding, stimulation of phosphorylation, and
Alzheimer’s disease, Neuron 3 (1989) 519–526. filament assembly depend on the degree of
43. Goedert M, Spillantini MG, Potier MC, Ulrich J, sulfation, J Biol Chem 272 (1997) 33118–33124.
Crowther RA, Cloning and sequencing of the 54. Hasegawa M, Jakes R, Crowther RA, Lee VM,
cDNA encoding an isoform of microtubule- Ihara Y, Goedert M, Characterization of mAb
associated protein tau containing four tandem AP422, a novel phosphorylation-dependent
repeats: differential expression of tau protein monoclonal antibody against tau protein, FEBS
mRNAs in human brain, EMBO J 8 (1989) 393– Lett 384 (1996) 25–30.
399. 55. Hasegawa M, Watanabe A, Takio K, Suzuki M,
44. Golombowski S, Muller-Spahn F, Romig H, Arai T, Titani K, Ihara Y, Characterization of two
Mendla K, Hock C, Dependence of cerebrospinal distinct monoclonal antibodies to paired helical
fluid Tau protein levels on apolipoprotein E4 filaments: further evidence for fetal-type
allele frequency in patients with Alzheimer’s phosphorylation of the tau in paired helical
disease, Neurosci Lett 225 (1997) 213–215. filaments, J Neurochem 60 (1993) 2068–2077.
45. Gong CX, Shaikh S, Wang JZ, Zaidi T, Grundke- 56. Heutink P, Stevens M, Rizzu P, Bakker E, Kros
Iqbal I, Iqbal K, Phosphatase activity toward JM, Tibben A, Niermeijer MF, van Duijn CM,
abnormally phosphorylated tau: decrease in Oostra BA, van Swieten JC, Hereditary fronto-
Alzheimer disease brain, J Neurochem 65 (1995) temporal dementia is linked to chromosome
732–738. 17q21-q22: a genetic and clinicopathological
46. Gong CX, Singh TJ, Grundke-Iqbal I, Iqbal K, study of three Dutch families, Ann Neurol 41
Phosphoprotein phosphatase activities in (1997) 150–159.
Alzheimer disease brain, J Neurochem 61 (1993) 57. Higgins JJ, Litvan I, Pho LT, Li W, Nee LE,
921–927. Progressive supranuclear gaze palsy is in linkage
47. Goode BL, Denis PE, Panda D, Radeke MJ, disequilibrium with the tau and not the alpha-
Miller HP, Wilson L, Feinstein SC, Functional synuclein gene, Neurology 50 (1998) 270–273.
interactions between the proline-rich and repeat 58. Hoffmann R, Lee VMY, Leight S, Varga I, Otvos
regions of tau enhance microtubule binding and L Jr., Unique Alzheimer’s disease paired helical
assembly, Mol Biol Cell 8 (1997) 353–365. filament specific epitopes involve double
48. Gu Y, Oyama F, Ihara Y, Tau is widely phosphorylation at specific sites, Biochemistry
expressed in rat tissues, J Neurochem 67 (1996) 36 (1997) 8114–8124.
1235–1244. 59. Hong M, Chen DC, Klein PS, Lee VM, Lithium
49. Gustke N, Trinczek B, Biernat J, Mandelkow reduces tau phosphorylation by inhibition of
EM, Mandelkow E, Domains of tau protein and glycogen synthase kinase-3, J Biol Chem 272
interactions with microtubules, Biochemistry 33 (1997) 25326–25332.
(1994) 9511–9522. 60. Hong M, Lee VM, Insulin and insulin-like
50. Hall GF, Yao J, Lee G, Human tau becomes growth factor-1 regulate tau phosphorylation in
phosphorylated and forms filamentous deposits cultured human neurons, J Biol Chem 272 (1997)
346 G.V.W. Johnson and J.A. Hartigan / Tau Protein in Normal and Alzheimer’s Disease Brain

19547–19553. 67–71.
61. Hoshi M, Takashima A, Noguchi K, Murayama 70. Jensen M, Basun H, Lannfelt L, Increased
M, Sato M, Kondo S, Saitoh Y, Ishiguro K, cerebrospinal fluid tau in patients with
Hoshino T, Imahori K, Regulation of Alzheimer’s disease, Neurosci Lett 186 (1995)
mitochondrial pyruvate dehydrogenase activity 189–191.
by tau protein kinase I/glycogen synthase kinase 71. Jicha GA, Bowser R, Kazam IG, Davies P, Alz-
3beta in brain, Proc Natl Acad Sci USA 93 50 and MC-1, a new monoclonal antibody raised
(1996) 2719–2723. to paired helical filaments, recognize
62. Hutton M, Lendon CL, Rizzu P, Baker M, conformational epitopes on recombinant tau, J
Froelich S, Houlden H, Pickering-Brown S, Neurosci Res 48 (1997) 128–132.
Chakraverty S, Isaacs A, Grover A, Hackett J, 72. Jicha GA, Lane E, Vincent I, Otvos L, Jr,
Adamson J, Lincoln S, Dickson D, Davies P, Hoffmann R, Davies P, A conformation- and
Petersen RC, Stevens M, de Graaff E, Wauters E, phosphorylation-dependent antibody recognizing
van Baren J, Hillebrand M, Joosse M, Kwon JM, the paired helical filaments of Alzheimer’s
Nowotny P, Che LK, Norton J, Morris JC, Reed disease, J Neurochem 69 (1997) 2087–2095.
LA, Trojanowski J, Basun H, Lannfelt L, Neystat 73. Johnson GV, Seubert P, Cox TM, Motter R,
M, Fahn S, Dark F, Tannenberg T, Dodd PR, Brown JP, Galasko D, The tau protein in human
Hayward N, Kwok JBJ, Schofield PR, Andreadis cerebrospinal fluid in Alzheimer’s disease
A, Snowden J, Craufurd D, Neary D, Owen F, consists of proteolytically derived fragments, J
Oostra BA, Hardy J, Goate A, van Swieten J, Neurochem 68 (1997) 430–433.
Mann D, Lynch T, Heutink P, Association of 74. Johnson GVW, Jenkins SM, Tau protein in
missense and 5'-splice-site mutations in tau with normal and Alzheimer’s disease brain,
the inherited dementia FTDP-17, Nature 393 Alzheimer’s Disease Rev 1 (1996) 38–54.
(1998) 702–705. 75. Johnson GVW, Litersky JM, Phosphorylation of
63. Hwang SC, Jhon DY, Bae YS, Kim JH, Rhee tau protein, Recent Res Devel in Neurochem 1
SG, Activation of phospholipase C-gamma by the (1996) 89–98.
concerted action of tau proteins and arachidonic 76. Kampers T, Friedhoff P, Biernat J, Mandelkow
acid, J Biol Chem 271 (1996) 18342–18349. EM, Mandelkow E, RNA stimulates aggregation
64. Illenberger S, Zheng-Fischhofer Q, Preuss U, of microtubule-associated protein tau into
Stamer K, Baumann K, Trinczek B, Biernat J, Alzheimer-like paired helical filaments, FEBS
Godemann R, Mandelkow EM, Mandelkow E, Lett 399 (1996) 344–349.
The endogenous and cell cycle-dependent 77. Kanemaru K, Takio K, Miura R, Titani K, Ihara
phosphorylation of tau protein in living cells: Y, Fetal-type phosphorylation of the tau in paired
Implications for Alzheimer’s Disease, Mol Biol helical filaments, J Neurochem 58 (1992) 1667–
Cell 9 (1998) 1495–1512. 1675.
65. Irving NG, Miller CC, Tau phosphorylation in 78. Kato T, Sasaki H, Katagiri T, Koiwai K, Youki
cells transfected with wild-type or an H, Totsuka S, Ishii T, The binding of basic
Alzheimer’s disease mutant Presenilin 1, fibroblast growth factor to Alzheimer’s
Neurosci Lett 222 (1997) 71–74. neurofibrillary tangles and senile plaques,
66. Ishiguro K, Takamatsu M, Tomizawa K, Omori Neurosci Lett 122 (1991) 33–36.
A, Takahashi M, Arioka M, Uchida T, Imahori 79. Kempf M, Clement A, Faissner A, Lee G, Brandt
K, Tau protein kinase I converts normal tau R, Tau binds to the distal axon early in
protein into A68-like component of paired helical development of polarity in a microtubule- and
filaments, J Biol Chem 267 (1992) 10897–10901. microfilament-dependent manner, J Neurosci 16
67. Islam K, Levy E, Carboxyl-terminal fragments of (1996) 5583–5592.
beta-amyloid precursor protein bind to 80. Kimura T, Ono T, Takamatsu J, Yamamoto H,
microtubules and the associated protein tau, Am J Ikegami K, Kondo A, Hasegawa M, Ihara Y,
Pathol 151 (1997) 265–271. Miyamoto E, Miyakawa T, Sequential changes of
68. Jancsik V, Filliol D, Rendon A, Tau proteins bind tau-site-specific phosphorylation during
to kinesin and modulate its activation by development of paired helical filaments,
microtubules, Neurobiology 4 (1996) 417–429. Dementia 7 (1996) 177–181.
69. Jenkins SM, Johnson GVW, Tau complexes with 81. Klein PS, Melton DA, A molecular mechanism
phospholipase C-γ in situ, Neuroreport 9 (1998) for the effect of lithium on development, Proc
G.V.W. Johnson and J.A. Hartigan / Tau Protein in Normal and Alzheimer’s Disease Brain 347

Natl Acad Sci USA 93 (1996) 8455–8459. effects on the organization and stability of
82. Ko L, Odawara T, Yen SH, Menadione-induced microtubules, Neuroscience 73 (1996) 1145–
tau dephosphorylation in cultured human 1157.
neuroblastoma cells, Brain Res 760 (1997) 118– 93. Mandelkow EM, Drewes G, Biernat J, Gustke N,
128. Van Lint J, Vandenheede JR, Mandelkow E,
83. Kondratick CM, Vandre DD, Alzheimer’s Glycogen synthase kinase-3 and the Alzheimer-
disease neurofibrillary tangles contain mitosis- like state of microtubule-associated protein tau,
specific phosphoepitopes, J Neurochem 67 FEBS Lett 314 (1992) 315–321.
(1996) 2405–2416. 94. Mandelkow EM, Mandelkow E, Tau as a marker
84. Ksiezak-Reding H, Tracz E, Yang LS, Dickson for Alzheimer’s disease, Trends Biochem Sci 18
DW, Simon M, Wall JS, Ultrastructural (1993) 480–483.
instability of paired helical filaments from 95. Mandell JW, Banker GA, A spatial gradient of
corticobasal degeneration as examined by tau protein phosphorylation in nascent axons, J
scanning transmission electron microscopy, Am J Neurosci 16 (1996) 5727–5740.
Pathol 149 (1996) 639–651. 96. Mattson MP, Cellular actions of beta-amyloid
85. Kuret J, Johnson GS, Cha D, Christenson ER, precursor protein and its soluble and fibrillogenic
DeMaggio AJ, Hoekstra MF, Casein kinase 1 is derivatives, Physiol Rev 77 (1997) 1081–1132.
tightly associated with paired-helical filaments 97. Mattson MP, Cheng B, Davis D, Bryant K,
isolated from Alzheimer’s disease brain, J Lieberburg I, Rydel RE, beta-Amyloid peptides
Neurochem 69 (1997) 2506–2515. destabilize calcium homeostasis and render
86. Lannfelt L, Basun H, Wahlund LO, Rowe BA, human cortical neurons vulnerable to
Wagner SL, Decreased alpha-secretase-cleaved excitotoxicity, J Neurosci 12 (1992) 376–89.
amyloid precursor protein as a diagnostic marker 98. Mattson MP, Fu W, Waeg G, Uchida K, 4-
for Alzheimer’s disease, Nat Med 1 (1995) 829– Hydroxynonenal, a product of lipid peroxidation,
832. inhibits dephosphorylation of the microtubule-
87. Le WD, Xie WJ, Kong R, Appel SH, Beta- associated protein tau, Neuroreport 8 (1997)
amyloid-induced neurotoxicity of a hybrid septal 2275–2281.
cell line associated with increased tau 99. Mattson MP, Tomaselli KJ, Rydel RE, Calcium-
phosphorylation and expression of beta-amyloid destabilizing and neurodegenerative effects of
precursor protein, J Neurochem 69 (1997) 978– aggregated beta-amyloid peptide are attenuated
985. by basic FGF, Brain Res 621 (1993) 35–49.
88. Lee VM, Balin BJ, Otvos L, Jr, Trojanowski JQ, 100. Mawal-Dewan M, Henley J, Van de Voorde A,
A68: a major subunit of paired helical filaments Trojanowski JQ, Lee VM, The phosphorylation
and derivatized forms of normal Tau, Science state of tau in the developing rat brain is
251 (1991) 675–678. regulated by phosphoprotein phosphatases, J Biol
89. Leger J, Kempf M, Lee G, Brandt R, Conversion Chem 269 (1994) 30981–30987.
of serine to aspartate imitates phosphorylation- 101. Mawal-Dewan M, Schmidt ML, Balin B, Perl
induced changes in the structure and function of DP, Lee VM, Trojanowski JQ, Identification of
microtubule-associated protein tau, J Biol Chem phosphorylation sites in PHF-TAU from patients
272 (1997) 8441–8446. with Guam amyotrophic lateral sclerosis/
90. Lindh M, Blomberg M, Jensen M, Basun H, parkinsonism-dementia complex, J Neuropathol
Lannfelt L, Engvall B, Scharnagel H, Marz W, Exp Neurol 55 (1996) 1051–1059.
Wahlund LO, Cowburn RF, Cerebrospinal fluid 102. McDermott JB, Aamodt S, Aamodt E, ptl-1, a
apolipoprotein E (apoE) levels in Alzheimer’s Caenorhabditis elegans gene whose products are
disease patients are increased at follow up and homologous to the tau microtubule-associated
show a correlation with levels of tau protein, proteins, Biochemistry 35 (1996) 9415–9423.
Neurosci Lett 229 (1997) 85–88. 103. Merrick SE, Demoise DC, Lee VM, Site-specific
91. Litersky JM, Johnson GV, Phosphorylation of tau dephosphorylation of tau protein at
in situ: inhibition of calcium-dependent Ser202/Thr205 in response to microtubule
proteolysis, J Neurochem 65 (1995) 903–911. depolymerization in cultured human neurons
92. Lovestone S, Hartley CL, Pearce J, Anderton BH, involves protein phosphatase 2A, J Biol Chem
Phosphorylation of tau by glycogen synthase 271 (1996) 5589–5594.
kinase-3 beta in intact mammalian cells: the 104. Michaelis ML, Ranciat N, Chen Y, Bechtel M,
348 G.V.W. Johnson and J.A. Hartigan / Tau Protein in Normal and Alzheimer’s Disease Brain

Ragan R, Hepperle M, Liu Y, Georg G, of apolipoprotein E genotype, Ann Neurol 37


Protection against b-amyloid toxicity in primary (1995) 512–518.
neurons by paclitaxel (Taxol), J Neurochem 70 115. Otto M, Wiltfang J, Tumani H, Zerr I, Lantsch
(1998) 1623–1627. M, Kornhuber J, Weber T, Kretzschmar HA,
105. Montine TJ, Amarnath V, Martin ME, Poser S, Elevated levels of tau-protein in
Strittmatter WJ, Graham DG, E-4-hydroxy-2- cerebrospinal fluid of patients with Creutzfeldt-
nonenal is cytotoxic and cross-links cytoskeletal Jakob disease, Neurosci Lett 225 (1997) 210–
proteins in P19 neuroglial cultures, Am J Pathol 212.
148 (1996) 89–93. 116. Otvos L Jr., Feiner L, Lang E, Szendrei GI,
106. Mori H, Hosoda K, Matsubara E, Nakamoto T, Goedert M, Lee VM, Monoclonal antibody PHF-
Furiya Y, Endoh R, Usami M, Shoji M, 1 recognizes tau protein phosphorylated at serine
Maruyama S, Hirai S, Tau in cerebrospinal residues 396 and 404, J Neurosci Res 39 (1994)
fluids: establishment of the sandwich ELISA 669–673.
with antibody specific to the repeat sequence in 117. Paudel HK, The regulatory Ser262 of micro-
tau, Neurosci Lett 186 (1995) 181–183. tubule-associated protein tau is phosphorylated
107. Morishima-Kawashima M, Hasegawa M, Takio by phosphorylase kinase, J Biol Chem 272
K, Suzuki M, Yoshida H, Titani K, Ihara Y, (1997) 1777–1785.
Proline-directed and non-proline-directed 118. Pei JJ, Tanaka T, Tung YC, Braak E, Iqbal K,
phosphorylation of PHF-tau, J Biol Chem 270 Grundke-Iqbal I, Distribution, levels, and activity
(1995) 823–829. of glycogen synthase kinase-3 in the Alzheimer
108. Motter R, Vigo-Pelfrey C, Kholodenko D, disease brain, J Neuropathol Exp Neurol 56
Barbour R, Johnson-Wood K, Galasko D, Chang (1997) 70–78.
L, Miller B, Clark C, Green R, et al. Reduction of 119. Perez M, Valpuesta JM, Medina M, Montejo de
beta-amyloid peptide42 in the cerebrospinal fluid Garcini E, Avila J, Polymerization of tau into
of patients with Alzheimer’s disease, Ann Neurol filaments in the presence of heparin: the minimal
38 (1995) 643–648. sequence required for tau-tau interaction, J
109. Munoz-Montano JR, Moreno FJ, Avila J, Diaz- Neurochem 67 (1996) 1183–1190.
Nido J, Lithium inhibits Alzheimer’s disease-like 120. Perry G, Siedlak SL, Richey P, Kawai M, Cras P,
tau protein phosphorylation in neurons, FEBS Kalaria RN, Galloway PG, Scardina JM, Cordell
Lett 411 (1997) 183–188. B, Greenberg BD, et al. Association of heparan
110. Munroe WA, Southwick PC, Chang L, Scharre sulfate proteoglycan with the neurofibrillary
DW, Echols CL Jr., Fu PC, Whaley JM, Wolfert tangles of Alzheimer’s disease, J Neurosci 11
RL, Tau protein in cerebrospinal fluid as an aid (1991) 3679–3683.
in the diagnosis of Alzheimer’s disease, Ann Clin 121. Peskind ER, Leverenz J, Farlow MR, Ito RK,
Lab Sci 25 (1995) 207–217. Provow SA, Siegel RS, Cleveland M, Morgan
111. Murrell JR, Koller D, Foroud T, Goedert M, CH, Pandian MR, Corbin S, Nochlin D,
Spillantini MG, Edenberg HJ, Farlow MR, Ghetti Schellenberg GD, Raskind MA, Wagner SL,
B, Familial multiple-system tauopathy with Clinicopathologic correlations of soluble amyloid
presenile dementia is localized to chromosome beta-protein precursor in cerebrospinal fluid in
17, Am J Hum Genet 61 (1997) 1131–1148. patients with Alzheimer disease and controls,
112. Nacharaju P, Ko L, Yen SH, Characterization of Alzheimer Dis Assoc Disord 11 (1997) 201–206.
in vitro glycation sites of tau, J Neurochem 69 122. Poorkaj P, Bird TD, Wijsman E, Nemens E,
(1997) 1709–1719. Garruto RM, Anderson L, Andreadis A,
113. Nelson PT, Stefansson K, Gulcher J, Saper CB, Wiederholt WC, Raskind M, Schellenberg GD,
Molecular evolution of tau protein: Implications Tau is a candidate gene for chromosome 17
for Alzheimer’s disease, J Neurochem 67 (1996) frontotemporal dementia, Ann Neurol 43 (1998)
1622–1632. 815–825.
114. Nitsch RM, Rebeck GW, Deng M, Richardson 123. Preuss U, Biernat J, Mandelkow EM, Mandelkow
UI, Tennis M, Schenk DB, Vigo-Pelfrey C, E, The ‘jaws’ model of tau-microtubule
Lieberburg I, Wurtman RJ, Hyman BT, et al. interaction examined in CHO cells, J Cell Sci 110
Cerebrospinal fluid levels of amyloid beta- (1997) 789–800.
protein in Alzheimer’s disease: inverse 124. Reed LA, Grabowski TJ, Schmidt ML, Morris
correlation with severity of dementia and effect JC, Goate A, Solodkin A, Van Hoesen GW,
G.V.W. Johnson and J.A. Hartigan / Tau Protein in Normal and Alzheimer’s Disease Brain 349

Schelper RL, Talbot CJ, Wragg MA, Mandelkow E, Oxidation of cysteine-322 in the
Trojanowski JQ, Autosomal dominant dementia repeat domain of microtubule-associated protein
with widespread neurofibrillary tangles [see tau controls the in vitro assembly of paired
comments], Ann Neurol 42 (1997) 564–572. helical filaments, Proc Natl Acad Sci USA 92
125. Reynolds CH, Nebreda AR, Gibb GM, Utton (1995) 8463–8467.
MA, Anderton BH, Reactivating kinase/p38 135. Scott CW, Spreen RC, Herman JL, Chow FP,
phosphorylates tau protein in vitro, J Neurochem Davison MD, Young J, Caputo CB,
69 (1997) 191–198. Phosphorylation of recombinant tau by cAMP-
126. Reynolds CH, Utton MA, Gibb GM, Yates A, dependent protein kinase, Identification of
Anderton BH, Stress-activated protein kinase/c- phosphorylation sites and effect on microtubule
jun N-terminal kinase phosphorylates tau protein, assembly, J Biol Chem 268 (1993) 1166–1173.
J Neurochem 68 (1997) 1736–1744. 136. Seidl R, Schuller E, Cairns N, Lubec G, Evidence
127. Riemenschneider M, Buch K, Schmolke M, Kurz against increased glycoxidation in patients with
A, Guder WG, Cerebrospinal protein tau is Alzheimer’s disease, Neurosci Lett 232 (1997)
elevated in early Alzheimer’s disease, Neurosci 49–52.
Lett 212 (1996) 209–211. 137. Selkoe DJ, Normal and abnormal biology of the
128. Sadot E, Gurwitz D, Barg J, Behar L, Ginzburg I, beta-amyloid precursor protein, Annu Rev
Fisher A, Activation of m1 muscarinic acetyl- Neurosci 17 (1994) 489–517.
choline receptor regulates tau phosphorylation in 138. Selkoe DJ, Abraham C, Ihara Y, Brain
transfected PC12 cells, J Neurochem 66 (1996) transglutaminase: in vitro crosslinking of human
877–880. neurofilament proteins into insoluble polymers,
129. Sato-Harada R, Okabe S, Umeyama T, Kanai Y, Proc Natl Acad Sci USA 79 (1982) 6070–6074.
Hirokawa N, Microtubule-associated proteins 139. Sengupta A, Wu Q, Grundke-Iqbal I, Iqbal K,
regulate microtubule function as the track for Singh TJ, Potentiation of GSK-3-catalyzed
intracellular membrane organelle transports, Cell Alzheimer-like phosphorylation of human tau by
Struct Funct 21 (1996) 283–295. cdk5, Mol Cell Biochem 167 (1997) 99–105.
130. Saunders AM, Schmader K, Breitner JC, Benson 140. Sergeant N, David JP, Goedert M, Jakes R,
MD, Brown WT, Goldfarb L, Goldgaber D, Vermersch P, Buee L, Lefranc D, Wattez A,
Manwaring MG, Szymanski MH, McCown N, et Delacourte A, Two-dimensional characterization
al. Apolipoprotein E epsilon 4 allele distributions of paired helical filament-tau from Alzheimer’s
in late-onset Alzheimer’s disease and in other disease: demonstration of an additional 74-kDa
amyloid-forming diseases [see comments], component and age-related biochemical
Lancet 342 (1993) 710–711. modifications, J Neurochem 69 (1997) 834–844.
131. Schmidt ML, DiDario AG, Lee VM, Trojanowski 141. Seubert P, Mawal-Dewan M, Barbour R, Jakes R,
JQ, An extensive network of PHF tau-rich Goedert M, Johnson GV, Litersky JM, Schenk D,
dystrophic neurites permeates neocortex and Lieberburg I, Trojanowski JQ, et al. Detection of
nearly all neuritic and diffuse amyloid plaques in phosphorylated Ser262 in fetal tau, adult tau, and
Alzheimer disease, FEBS Lett 344 (1994) 69–73. paired helical filament tau, J Biol Chem 270
132. Schmidt ML, Huang R, Martin JA, Henley J, (1995) 18917–18922.
Mawal-Dewan M, Hurtig HI, Lee VM, 142. Shea TB, Prabhakar S, Ekinci FJ, Beta-amyloid
Trojanowski JQ, Neurofibrillary tangles in and ionophore A23187 evoke tau
progressive supranuclear palsy contain the same hyperphosphorylation by distinct intracellular
tau epitopes identified in Alzheimer’s disease pathways: differential involvement of the
PHFtau, J Neuropathol Exp Neurol 55 (1996) calpain/protein kinase C system, J Neurosci Res
534–539. 49 (1997) 759–768.
133. Schroder J, Pantel J, Ida N, Essig M, Hartmann 143. Shiurba RA, Ishiguro K, Takahashi M, Sato K,
T, Knopp MV, Schad LR, Sandbrink R, Sauer H, Spooner ET, Mercken M, Yoshida R, Wheelock
Masters CL, Beyreuther K, Cerebral changes and TR, Yanagawa H, Imahori K, Nixon RA,
cerebrospinal fluid beta-amyloid in Alzheimer’s Immunocytochemistry of tau phosphoserine 413
disease: a study with quantitative magnetic and tau protein kinase I in Alzheimer pathology,
resonance imaging, Mol Psychiatry 2 (1997) Brain Res 737 (1996) 119–132.
505–507. 144. Snow AD, Lara S, Nochlin D, Wight TN,
134. Schweers O, Mandelkow EM, Biernat J, Cationic dyes reveal proteoglycans structurally
350 G.V.W. Johnson and J.A. Hartigan / Tau Protein in Normal and Alzheimer’s Disease Brain

integrated within the characteristic lesions of 154. Szendrei GI, Lee VM, Otvos L Jr., Recognition
Alzheimer’s disease, Acta Neuropathol 78 (1989) of the minimal epitope of monoclonal antibody
113–123. Tau-1 depends upon the presence of a phosphate
145. Snow AD, Mar H, Nochlin D, Sekiguchi RT, group but not its location, J Neurosci Res 34
Kimata K, Koike Y, Wight TN, Early (1993) 243–249.
accumulation of heparan sulfate in neurons and in 155. Takashima A, Noguchi K, Michel G, Mercken
the beta-amyloid protein-containing lesions of M, Hoshi M, Ishiguro K, Imahori K, Exposure of
Alzheimer’s disease and Down’s syndrome, Am rat hippocampal neurons to amyloid beta peptide
J Pathol 137 (1990) 1253–1270. (25-35) induces the inactivation of phosphatidyl
146. Sontag E, Nunbhakdi-Craig V, Bloom GS, inositol-3 kinase and the activation of tau protein
Mumby MC, A novel pool of protein kinase I/glycogen synthase kinase-3 beta,
phosphatase 2A is associated with microtubules Neurosci Lett 203 (1996) 33–36.
and is regulated during the cell cycle, J Cell Biol 156. Takashima A, Yamaguchi H, Noguchi K, Michel
128 (1995) 1131–1144. G, Ishiguro K, Sato K, Hoshino T, Hoshi M,
147. Sontag E, Nunbhakdi-Craig V, Lee G, Bloom Imahori K, Amyloid beta peptide induces
GS, Mumby MC, Regulation of the cytoplasmic accumulation of amyloid protein
phosphorylation state and microtubule-binding precursor via tau protein kinase I/glycogen
activity of Tau by protein phosphatase 2A, synthase kinase-3 beta in rat hippocampal
Neuron 17 (1996) 1201–1207. neurons, Neurosci Lett 198 (1995) 83–86.
148. Spillantini MG, Crowther RA, Goedert M, 157. Tamaoka A, Sawamura N, Fukushima T, Shoji S,
Comparison of the neurofibrillary pathology in Matsubara E, Shoji M, Hirai S, Furiya Y, Endoh
Alzheimer’s disease and familial presenile R, Mori H, Amyloid beta protein 42(43) in
dementia with tangles, Acta Neuropathol 92 cerebrospinal fluid of patients with Alzheimer’s
(1996) 42–48. disease, J Neurol Sci 148 (1997) 41–45.
149. Spillantini MG, Goedert M, Crowther RA, 158. Tato RE, Frank A, Hernanz A, Tau protein
Murrell JR, Farlow MR, Ghetti B, Familial concentrations in cerebrospinal fluid of patients
multiple system tauopathy with presenile with dementia of the Alzheimer type, J Neurol
dementia: a disease with abundant neuronal and Neurosurg Psychiatry 59 (1995) 280–283.
glial tau filaments, Proc Natl Acad Sci USA 94 159. Thurston VC, Zinkowski RP, Binder LI, Tau as a
(1997) 4113–4118. nucleolar protein in human nonneural cells in
150. Spillantini MG, Murrell JR, Goedert M, Farlow vitro and in vivo, Chromosoma 105 (1996) 20–
MR, Klug A, Ghetti B, Mutation in the tau gene 30.
in familial multiple system tauopathy with 160. Trinczek B, Biernat J, Baumann K, Mandelkow
presenile dementia, Proc Natl Acad Sci USA 95 EM, Mandelkow E, Domains of tau protein,
(1998) 7737–7741. differential phosphorylation, and dynamic
151. Stambolic V, Ruel L, Woodgett JR, Lithium instability of microtubules, Mol Biol Cell 6
inhibits glycogen synthase kinase-3 activity and (1995) 1887–1902.
mimics wingless signalling in intact cells 161. Utton MA, Vandecandelaere A, Wagner U,
[published erratum appears in Curr Biol 1997 Reynolds CH, Gibb GM, Miller CC, Bayley PM,
Mar 1 7(3) 196], Curr Biol 6 (1996) 1664–1668. Anderton BH, Phosphorylation of tau by
152. Sturchler-Pierrat C, Abramowski D, Duke M, glycogen synthase kinase 3beta affects the ability
Wiederhold KH, Mistl C, Rothacher S, of tau to promote microtubule self-assembly,
Ledermann B, Burki K, Frey P, Paganetti PA, Biochem J 323 (1997) 741–747.
Waridel C, Calhoun ME, Jucker M, Probst A, 162. Vandermeeren M, Mercken M, Vanmechelen E,
Staufenbiel M, Sommer B, Two amyloid Six J, van de Voorde A, Martin JJ, Cras P,
precursor protein transgenic mouse models with Detection of tau proteins in normal and
Alzheimer disease-like pathology, Proc Natl Alzheimer’s disease cerebrospinal fluid with a
Acad Sci USA 94 (1997) 13287–13292. sensitive sandwich enzyme-linked immuno-
153. Su JH, Cummings BJ, Cotman CW, Plaque sorbent assay, J Neurochem 61 (1993) 1828–
biogenesis in brain aging and Alzheimer’s 1834.
disease. I. Progressive changes in phos- 163. Vandre DD, Centonze VE, Peloquin J, Tombes
phorylation states of paired helical filaments and RM, Borisy GG, Proteins of the mammalian
neurofilaments, Brain Res 739 (1996) 79–87. mitotic spindle: phosphorylation/dephosphoryl-
G.V.W. Johnson and J.A. Hartigan / Tau Protein in Normal and Alzheimer’s Disease Brain 351

ation of MAP-4 during mitosis, J Cell Sci 98 Pathol 150 (1997) 2181–2195.
(1991) 577–588. 171. Wischik CM, Edwards PC, Lai RY, Roth M,
164. Vigo-Pelfrey C, Seubert P, Barbour R, Blomquist Harrington CR, Selective inhibition of Alzheimer
C, Lee M, Lee D, Coria F, Chang L, Miller B, disease-like tau aggregation by phenothiazines,
Lieberburg I, et al. Elevation of microtubule- Proc Natl Acad Sci USA 93 (1996) 11213–
associated protein tau in the cerebrospinal fluid 11218.
of patients with Alzheimer’s disease, Neurology 172. Wu JM, Chen Y, Hsieh TC, Brandt R, Lee G,
45 (1995) 788–793. Phosphorylation of native and truncated isoforms
165. Vincent I, Jicha G, Rosado M, Dickson DW, of protein tau by the double-stranded DNA-
Aberrant expression of mitotic cdc2/cyclin B1 dependent protein kinase (DNA-PK) shows that
kinase in degenerating neurons of Alzheimer’s the primary phosphorylation sites are localized
disease brain, J Neurosci 17 (1997) 3588–3598. between amino acid residues 212–231 of the
166. Vincent I, Rosado M, Davies P, Mitotic longest tau, Biochem Mol Biol Int 40 (1996) 21–
mechanisms in Alzheimer’s disease? J Cell Biol 31.
132 (1996) 413–425. 173. Xie HQ, Johnson GVW, Calcineurin inhibition
167. Wagner U, Utton M, Gallo JM, Miller CC, prevents calpain-mediated proteolysis of tau in
Cellular phosphorylation of tau by GSK-3 beta differentiated PC12 cells, J Neurosci Res 53
influences tau binding to microtubules and (1998) 153–164.
microtubule organisation, J Cell Sci 109 (1996) 174. Xie HQ, Litersky JM, Hartigan JA, Jope RS,
1537–1543. Johnson GVW, The interrelationship between
168. Wang JZ, Grundke-Iqbal I, Iqbal K, selective tau phosphorylation and microtubule
Glycosylation of microtubule-associated protein association, Brain Res 798 (1998) 173–183.
tau: an abnormal posttranslational modification in 175. Zemlan FP, Dean GE, Monoclonal antibody
Alzheimer’s disease [see comments], Nat Med 2 PHF-9 recognizes phosphorylated serine 404 of
(1996) 871–875. tau protein and labels paired helical filaments, J
169. Watanabe A, Hasegawa M, Suzuki M, Takio K, Neurosci Res 46 (1996) 90–97.
Morishima-Kawashima M, Titani K, Arai T, 176. Zheng-Fischhofer Q, Biernat J, Mandelkow EM,
Kosik KS, Ihara Y, In vivo phosphorylation sites Illenberger S, Godemann R, Mandelkow E,
in fetal and adult rat tau, J Biol Chem 268 (1993) Sequential phosphorylation of Tau by glycogen
25712–25717. synthase kinase-3beta and protein kinase A at
170. Wilson DM, Binder LI, Free fatty acids stimulate Thr212 and Ser214 generates the Alzheimer-
the polymerization of tau and amyloid beta specific epitope of antibody AT100 and requires
peptides. In vitro evidence for a common effector a paired-helical-filament-like conformation, Eur J
of pathogenesis in Alzheimer’s disease, Am J Biochem 252 (1998) 542–552.

You might also like