You are on page 1of 41

P1: FUM

April 7, 2001 15:58 Annual Reviews AR121-37

Annu. Rev. Neurosci. 2001. 24:1121–159


Copyright °
c 2001 by Annual Reviews. All rights reserved

NEURODEGENERATIVE TAUOPATHIES
Virginia M-Y Lee,1 Michel Goedert,2
and John Q Trojanowski1
1Center for Neurodegenerative Disease Research, Department of Pathology and
Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia,
Annu. Rev. Neurosci. 2001.24:1121-1159. Downloaded from www.annualreviews.org

Pennsylvania 19104; e-mail: vmylee@mail.med.upenn.edu,


trojanow@mail.med.upenn.edu
2Medical Research Council, Laboratory of Molecular Biology, Hills Road, Cambridge,

CB2 2QH, United Kingdom; e-mail: mg@mrc-lmb.cam.ac.uk


by Open University on 07/11/13. For personal use only.

Key Words Alzheimer’s disease, frontotemporal dementia, mutation,


neurodegenerative disease, filamentous deposits, pathology, tau protein
■ Abstract The defining neuropathological characteristics of Alzheimer’s disease
are abundant filamentous tau lesions and deposits of fibrillar amyloid β peptides.
Prominent filamentous tau inclusions and brain degeneration in the absence of
β -amyloid deposits are also hallmarks of neurodegenerative tauopathies exemplified by
sporadic corticobasal degeneration, progressive supranuclear palsy, and Pick’s dis-
ease, as well as by hereditary frontotemporal dementia and parkinsonism linked to
chromosome 17 (FTDP-17). Because multiple tau gene mutations are pathogenic
for FTDP-17 and tau polymorphisms appear to be genetic risk factors for sporadic
progressive supranuclear palsy and corticobasal degeneration, tau abnormalities are
linked directly to the etiology and pathogenesis of neurodegenerative disease. Indeed,
emerging data support the hypothesis that different tau gene mutations are pathogenic
because they impair tau functions, promote tau fibrillization, or perturb tau gene splic-
ing, thereby leading to formation of biochemically and structurally distinct aggre-
gates of tau. Nonetheless, different members of the same kindred often exhibit diverse
FTDP-17 syndromes, which suggests that additional genetic or epigenetic factors influ-
ence the phenotypic manifestations of neurodegenerative tauopathies. Although these
and other hypothetical mechanisms of neurodegenerative tauopathies remain to be
tested and validated, transgenic models are increasingly available for this purpose,
and they will accelerate discovery of more effective therapies for neurodegenerative
tauopathies and related disorders, including Alzheimer’s disease.

INTRODUCTION
The study of sporadic and familial neurodegenerative diseases over the past decade
has led to the realization that many of these disorders are characterized by distinct
hallmark brain lesions that have in common the formation of filamentous de-
posits of abnormal brain proteins. Thus, a group of heterogeneous dementias and

0147-006X/01/0621-1121$14.00 1121
P1: FUM
April 7, 2001 15:58 Annual Reviews AR121-37

1122 LEE ¥ GOEDERT ¥ TROJANOWSKI

movement disorders that are characterized neuropathologically by prominent intra-


cellular accumulations of abnormal filaments formed by the microtubule-associated
protein tau appears to share common mechanisms of disease. They are collectively
known as neurodegenerative tauopathies (Table 1). Despite their diverse pheno-
typic manifestations, brain dysfunction and degeneration in tauopathies is linked
to the progressive accumulation of filamentous tau inclusions, and this, together
with the absence of other disease-specific neuropathological abnormalities, pro-
vided circumstantial evidence implicating abnormal tau in disease onset and/or
progression. However, this view remained unproven and highly controversial un-
til 1998, when multiple tau gene mutations were discovered in frontotemporal
Annu. Rev. Neurosci. 2001.24:1121-1159. Downloaded from www.annualreviews.org

dementia and parkinsonism linked to chromosome 17 (FTDP-17), thereby pro-


viding unequivocal evidence that tau abnormalities alone are sufficient to cause
neurodegenerative disease (Foster et al 1997, Hutton et al 1998, Poorkaj et al
1998, Spillantini et al 1998c). This seminal finding opened up new avenues for
by Open University on 07/11/13. For personal use only.

TABLE 1 Diseases with tau-based neurofibrillary pathology


Alzheimer’s disease
Amyotrophic lateral sclerosis/parkinsonism–dementia complexa
Argyrophilic grain dementiaa
Corticobasal degenerationa
Creutzfeldt-Jakob disease
Dementia pugilisticaa
Diffuse neurofibrillary tangles with calcificationa
Down’s syndrome
Frontotemporal dementia with parkinsonism linked to chromosome 17a
Gerstmann-Sträussler-Scheinker disease
Hallervorden-Spatz disease
Myotonic dystrophy
Niemann-Pick disease, type C
Non-Guamanian motor neuron disease with neurofibrillary tangles
Pick’s diseasea
Postencephalitic parkinsonism
Prion protein cerebral amyloid angiopathy
Progressive subcortical gliosisa
Progressive supranuclear palsya
Subacute sclerosing panencephalitis
Tangle only dementiaa
a
Diseases in which tau-positive neurofibrillary pathology are the most predominant neu-
ropathologic feature.
P1: FUM
April 7, 2001 15:58 Annual Reviews AR121-37

NEW INSIGHTS INTO TAUOPATHIES 1123

investigating the role of tau abnormalities in mechanisms of brain dysfunction and


degeneration.
This review is designed to integrate and interpret the remarkable recent advances
that have led to new insights into the mechanistic role that tau abnormalities play
in neurodegenerative disease. It begins with a brief summary of our current under-
standing of the human tau gene and the functions of the six alternatively spliced
tau isoforms that are expressed in the normal adult human brain. What is known
about the role of tau abnormalities in Alzheimer’s disease (AD) is considered next,
and this is followed by an overview of several prototypical, as well as some novel
sporadic, tauopathies. This information provides the context and perspective in
Annu. Rev. Neurosci. 2001.24:1121-1159. Downloaded from www.annualreviews.org

which to present an update on the increasing number of pathogenic tau gene mu-
tations, as well as a summary of the neuropathological and biochemical profiles of
the tau pathologies in FTDP-17. Finally, the review concludes with an update of
the current status of efforts to develop transgenic (TG) and other animal models
by Open University on 07/11/13. For personal use only.

of human neurodegenerative tauopathies.

STRUCTURE, FUNCTION, AND MOLECULAR


GENETICS OF TAU
Tau proteins are low Mr microtubule-associated proteins that are abundant in the
central nervous system (CNS), where they are expressed predominantly in axons.
They are also expressed in axons of peripheral nervous system neurons but are
barely detectable in CNS astrocytes and oligodendrocytes (Cleveland et al 1977b,
Binder et al 1985, Shin et al 1991, Couchie et al 1992, LoPresti et al 1995). Human
tau proteins are encoded by a single gene consisting of 16 exons on chromosome
17q21, and the CNS isoforms are generated by alternative mRNA splicing of 11 of
these exons (Neve et al 1986, Goedert et al 1988, Andreadis et al 1992) (Figure 1).
In adult human brain, alternative splicing of exons (E)2, E3, and E10 generates
six tau isoforms ranging from 352 to 441 amino acids in length (Figure 1), which
differ by the presence of either three (3R-tau) or four (4R-tau) carboxy-terminal
tandem repeat sequences of 31 or 32 amino acids each that are encoded by E9,
E10, E11, and E12 (Goedert et al 1989a,b; Andreadis et al 1992). Additionally,
the triplets of 3R-tau and 4R-tau isoforms differ as a result of alternative splicing
of E2 and E3 to generate tau isoforms without (0N) or with either 29 (1N) or 58
(2N) amino acid inserts of unknown functions (Goedert et al 1989b). In adult
human brain, the ratio of 3R-tau to 4R-tau isoforms is ∼1, but the 1N, 0N, and
2N tau isoforms comprise about 54%, 37%, and 9%, respectively, of total tau
(Goedert & Jakes 1990, Hong et al 1998). In addition, the alternative splicing of
tau is developmentally regulated such that only the shortest tau isoform (3R/0N) is
expressed in fetal brain, whereas all six isoforms appear in the postnatal period of
the human brain (Goedert et al 1989a). In the peripheral nervous system, inclusion
of E4a in the amino-terminal half results in the expression of higher Mr proteins
termed big tau (Georgieff et al 1991, Couchie et al 1992, Goedert et al 1992c).
P1: FUM
April 7, 2001 15:58 Annual Reviews AR121-37

1124 LEE ¥ GOEDERT ¥ TROJANOWSKI


Annu. Rev. Neurosci. 2001.24:1121-1159. Downloaded from www.annualreviews.org

Figure 1 Schematic representation of the human tau gene and the six central nervous system
by Open University on 07/11/13. For personal use only.

(CNS) tau isoforms generated by alternative mRNA splicing. The human tau gene contains 16
exons, including exon (E)0, which is part of the promoter. Alternative splicing of E2, E3, and E10
(gray boxes) produces the six tau isoforms. E6 and E8 (stippled boxes) are not transcribed in the
human CNS. E4a (striped box), which is also not transcribed in the human CNS, is expressed in
the peripheral nervous system, leading to the larger tau isoforms, termed big tau (see text). The
black bars depict the 18–amino acid microtubule binding repeats and are designated R1 to R4.
The relative sizes of the exons and introns are not drawn to scale.

Since its discovery >25 years ago, a number of well-defined functions of tau
protein have been discovered and extensively characterized (for review, see Buée
et al 2000). Most notably, tau binds to and stabilizes microtubules (MTs), in ad-
dition to promoting MT polymerization (Weingarten et al 1975, Cleveland et al
1977b). The MT binding domains of tau are localized to the carboxy-terminal half
of the molecule within the four MT binding motifs. These motifs are composed of
highly conserved 18–amino acid long binding elements separated by flexible, but
less conserved, interrepeat sequences that are 13–14 amino acids long (Himmler
et al 1989, Lee et al 1989, Butner & Kirschner 1991). The binding of tau to MTs
is a complex process mediated in part by a flexible array of weak MT binding sites
that are distributed throughout the MT binding domain delineated by these repeats
and their interrepeat sequences (Lee et al 1989, Butner & Kirschner 1991). In ad-
dition, sequences flanking the repeats contribute to microtubule binding (Gustke
et al 1994). 4R-tau isoforms are more efficient at promoting MT assembly and
have a greater MT binding affinity than do 3R-tau isoforms (Goedert & Jakes
1990, Butner & Kirschner 1991). It is interesting to note that the interrepeat se-
quence between the first and second MT binding repeats has more than twice
the binding affinity of any individual MT binding repeat (Goode & Feinstein
1994). This region is unique to 4R-tau and is believed to be responsible for the
higher MT binding affinity of 4R-tau isoforms compared with 3R-tau isoforms
(Goedert & Jakes 1990). Notably, because other microtubule-associated proteins
P1: FUM
April 7, 2001 15:58 Annual Reviews AR121-37

NEW INSIGHTS INTO TAUOPATHIES 1125

probably perform similar functions, it is possible they can compensate for a defi-
ciency or loss of tau, which may account for the report that tau knockout mice did
not have any overt phenotype (Harada et al 1994). However, a more recent study
has reported some subtle behavioral impairments in older knockout mice (Ikegami
et al 2000).

TAU PHOSPHORYLATION AND REGULATION


OF TAU FUNCTIONS
Annu. Rev. Neurosci. 2001.24:1121-1159. Downloaded from www.annualreviews.org

There are 79 potential serine (Ser) and threonine (Thr) phosphate acceptor residues
in the longest tau isoform, and phosphorylation at ∼30 of these sites has been
reported in normal tau proteins (reviewed in Billingsley & Kincaid 1997, Buée
et al 2000). Tau phosphorylation is developmentally regulated such that fetal tau is
by Open University on 07/11/13. For personal use only.

more highly phosphorylated in embryonic compared with adult CNS (Kanemaru


et al 1992, Bramblett et al 1993, Goedert et al 1993, Watanabe et al 1993), and
the degree of phosphorylation of all six tau isoforms decreases with age, probably
because of the activation of phosphatases (Mawal-Dewan et al 1994). The tau
phosphorylation sites are clustered in regions flanking the MT binding repeats,
and it is well established that increasing tau phosphorylation negatively regulates
MT binding (Drechsel et al 1992, Bramblett et al 1993, Yoshida & Ihara 1993,
Biernat et al 1993). The importance of individual sites in tau in regulating MT
binding has been debated for some time. For example, phosphorylation of Ser262,
which lies within the first MT binding domain, is thought to play a dominant role
in reducing the binding of tau to MTs (Biernat et al 1993). A similar role may be
considered for phosphorylation of Ser396, which is located adjacent to the carboxy-
terminal end of the fourth MT binding domain (Bramblett et al 1993), but other
data argue that phosphorylation of neither of these sites is sufficient to eliminate
the binding of tau to MTs (Seubert et al 1995). It is interesting that both sites are
phosphorylated in fetal tau and that they are hyperphosphorylated in all six tau
isoforms that form abnormal paired helical filaments (PHFs) in the neurofibrillary
tangles (NFTs) of the AD brain (Seubert et al 1995). The regulation of the binding
of tau to MTs may be also be regulated by sites outside the MT binding repeats.
For example, although the evidence is fragmentary, it has been suggested that a
heptapeptide sequence (224KKVAVVR230) located within a proline-rich domain,
amino-terminal to the MT binding domains, promotes MT binding in combination
with the repeat regions (Goode et al 1997). Thus, it is likely that phosphorylation at
multiple sites, especially those flanking the MT binding repeats, but also additional
intra- and intermolecular interactions, regulate the MT binding function of tau.
Only little is known about the regulation of tau phosphorylation. One study has
reported increased phosphorylation of tau at Ser202/Thr205 in mice that lack
Reelin or both the very-low-density lipoprotein receptor and the apolipoprotein
E receptor 2 (Hiesberger et al 1999). It suggests that tyrosine phsophorylation
of the disabled-1 adaptor protein may play a role in regulating the level of tau
P1: FUM
April 7, 2001 15:58 Annual Reviews AR121-37

1126 LEE ¥ GOEDERT ¥ TROJANOWSKI

phosphorylation. It remains to be seen whether this finding can be extended to


additional phosphorylation sites in tau and to mice mutant for other components
of this developmental pathway.
For the reasons discussed above, considerable research has focused on the
protein kinases and protein phosphatases that regulate tau phosphorylation, and a
large number of Ser/Thr protein kinases have been implicated or have been sug-
gested as playing a role in regulating tau functions in vivo (reviewed in Billingsley
& Kincaid 1997, Buée et al 2000, Hong et al 2000). These kinases include mito-
gen-activated protein kinase (Drewes et al 1992, Drechsel et al 1992, Goedert
et al 1992a), glycogen synthase kinase 3β (GSK-3β) (Hanger et al 1992,
Annu. Rev. Neurosci. 2001.24:1121-1159. Downloaded from www.annualreviews.org

Mandelkow et al 1992), cyclin-dependent kinase 2 (cdk2) (Baumann et al 1993),


cdk5 (Baumann et al 1993, Kobayashi et al 1993), cAMP-dependent protein ki-
nase (Litersky & Johnson 1992), Ca2+/calmodulin-dependent protein kinase II
(Baudier & Cole 1987), and MT-affinity regulating kinase (Drewes et al 1997).
by Open University on 07/11/13. For personal use only.

In addition, several members of the family of stress-activated protein kinases also


phosphorylate tau at multiple sites (Goedert et al 1997; Reynolds et al 1997a,b).
Nonetheless, it is important to emphasize that many of these studies provide only
in vitro evidence to implicate specific kinases and that it remains unclear what role
they play in the in vivo phosphorylation of tau.
Recent data have implicated two protein kinases, GSK-3β and cdk5, in the in
vivo regulation of tau phosphorylation. GSK-3β is a Ser/Thr kinase that is abundant
in brain and associates with MTs (Mandelkow et al 1992, Ishiguro et al 1994, Singh
et al 1995, Takahashi et al 1995, Cohen 1999). Cotransfection of nonneuronal
cells with human tau and GSK-3β induces hyperphosphorylation of tau associated
with a loss of MT binding (Lovestone et al 1996). In cultured neuronal cells,
GSK-3β-mediated phosphorylation of tau is inhibited by insulin and IGF-1 via a
phosphatidylinositol 3-kinase and protein kinase B–dependent signaling pathway
(Hong & Lee 1997). In addition, direct inhibition of GSK-3β by lithium salts or
ATP competitive inhibitors reduces tau phosphorylation and affects MT stability
(Hong et al 1997, Munoz-Montano et al 1997, Lovestone et al 1999, Takahashi
et al 1999, Leost et al 2000). Cdk5 is a Ser/Thr protein kinase highly enriched in
neurons that colocalizes to the cytoskeleton and contributes to the phosphorylation
of tau (Baumann et al 1993, Kobayashi et al 1993, Lew & Wang 1994). It is
activated by interaction with regulatory subunits, the best characterized of which
is p35 (Ishiguro et al 1994, Lew et al 1994, Tsai et al 1994). Recently, Sobue et al
(2000) demonstrated that cdk5 complexes with tau in a manner that depends on
the phosphorylation of tau, and that tau anchors cdk5 to MTs. Moreover, cdk5-
mediated tau phosphorylation stimulates further phosphorylation of tau by GSK-
3β (Yamaguchi et al 1996, Sengupta et al 1997). However, further work is needed
to determine the relative contributions of individual kinases to tau phosphorylation
in vivo.
Because protein phosphatases are required for counterbalancing the effects of
tau protein kinases, they have also been an intense focus of research. A number
of studies have implicated several phosphatases in regulating tau phosphorylation,
including PP1, PP2A, PP2B, and PP2C (reviewed in Billingsley & Kincaid 1997,
P1: FUM
April 7, 2001 15:58 Annual Reviews AR121-37

NEW INSIGHTS INTO TAUOPATHIES 1127

Buée et al 2000). They all dephosphorylate tau in vitro with overlapping speci-
ficities; however, their role in vivo is unclear. Both PP2A and PP2B are present in
human brain tissue, and they dephosphorylate tau in a site-specific manner. Both
enzymes dephosphorylate Ser396 (Matsuo et al 1994), whereas PP2A also dephos-
phorylates tau at multiple additional sites (Goedert et al 1992a, Drewes et al 1993,
Billingsley & Kincaid 1997). Of the phosphatase activity in rat brain, PP2A is the
major activity toward tau phosphorylated by a number of protein kinases (Goedert
et al 1992a, 1995a). PP1 and PP2A bind to tau, and this interaction is believed
to mediate an association with MTs (Sontag et al 1995, Liao et al 1998). PP2A
has also been demonstrated to bind directly to MTs, an interaction that regulates
Annu. Rev. Neurosci. 2001.24:1121-1159. Downloaded from www.annualreviews.org

its activity in vitro (Sontag et al 1999). Lastly, inhibition of PP1 and PP2A by
okadaic acid in cultured human NT2N neurons results in increased tau phospho-
rylation, accompanied by decreased tau binding to MTs, selective destruction of
stable MTs, and rapid degeneration of axons (Merrick et al 1997). As with the
by Open University on 07/11/13. For personal use only.

kinases implicated in the phosphorylation of tau, further studies are necessary to


define the specific role of individual phosphatases in the in vivo regulation of the
phosphorylation state of tau.

AD NEUROFIBRILLARY PATHOLOGY IS MADE OF


ABNORMALLY PHOSPHORYLATED TAU
Filamentous neuronal or neuronal and glial tau inclusions associated with the de-
generation of affected brain regions are the defining neuropathological features of
tauopathies. In AD, NFTs and neuropil thread pathology are found in conjunction
with the deposition of β-amyloid (Aβ) fibrils in the extracellular space. By light
microscopy, the neurofibrillary lesions of AD are stained with anti-tau antibod-
ies (Brion et al 1985, Grundke-Iqbal et al 1986). Ultrastructurally, the dominant
components of neurofibrillary lesions in AD are paired helical filaments (PHFs)
and straight filaments (Kidd 1963). PHFs are composed of two strands of filament
twisted around one another with a periodicity of 80 nm and a width varying from
8 to 20 nm (Crowther & Wischik 1985) whereas straight filaments lack this helical
periodicity (Crowther 1991). Both PHFs and straight filaments are composed pre-
dominantly of abnormally hyperphosphorylated tau proteins (Goedert et al 1988,
Kondo et al 1988, Kosik et al 1988, Wischik et al 1988, Lee et al 1991). Analysis
of PHFs purified from AD brains by sodium dodecyl sulphate–polyacrylamide
gel electrophoresis has revealed three major bands of approximately 68, 64, and
60 kDa, as well as a minor band of approximately 72 kDa (Greenberg & Davies
1990, Lee et al 1991) (Figure 2). Upon dephosphorylation, six bands are resolved
that correspond to the six isoforms of tau found in adult human brain (Lee et al
1991, Greenberg et al 1992, Goedert et al 1992b). The relative proportions of
the tau isoforms observed in AD PHFs are similar to those that are character-
istic of the six soluble tau isoforms observed in normal adult human brain (see
Trojanowski & Lee 1994, Morishima-Kawashima et al 1995, Goedert et al 1995b,
Hong et al 1998). Although many phosphorylation sites identified in PHFtau
P1: FUM
April 7, 2001 15:58 Annual Reviews AR121-37

1128 LEE ¥ GOEDERT ¥ TROJANOWSKI


Annu. Rev. Neurosci. 2001.24:1121-1159. Downloaded from www.annualreviews.org
by Open University on 07/11/13. For personal use only.

Figure 2 Schematic representation of Western blot banding patterns of insoluble and solu-
ble tau from different tauopathies. The cartoon depicts the typical banding pattern of nonde-
phosphorylated and dephosphorylated insoluble (filamentous) tau (top panels) as well as sol-
uble tau (bottom panels) from brains of patients with the diseases indicated following resolu-
tion by sodium dodecyl sulphate–polyacrylamide gel electrophoresis and immunoblotting with
phosphorylation-independent anti-tau antibodies. Nondephosphorylated insoluble tau from the
brain of patients with Alzheimer’s disease (AD) and some frontotemporal dementia and parkin-
sonism linked to chromosome 17 (FTDP-17), mutations that do not affect splicing (V337M and
R406W), runs as three major bands of 68, 64, and 60 kDa and as a minor, variable band of 72
kDa. When dephosphorylated, it resolves into six bands that correspond to soluble tau. In corti-
cobasal degeneration (CBD) and progressive supranuclear palsy (PSP), as well as FTDP-17 with
the P301L mutation, the two prominent 68- and 64-kDa insoluble tau bands are detected (the
72-kDa minor band is variably detected) and align with four tandem repeat sequences (4R-tau)
following dephosphorylation. The soluble fraction shows all six isoforms, indicating that there is
selective aggregation of 4R-tau. In contrast, in FTDP-17 mutations that affect mRNA splicing,
there is expression of predominantly soluble 4R-tau throughout the entire brain. Only 4R-tau is
deposited in a filamentous form. In Pick’s disease (PiD) and some FTDP-17 mutations (K257T,
G389R) that do not affect splicing, the lower two 64- and 60-kDa insoluble tau bands predomi-
nate. Following dephosphorylation, a predominance of 3R-tau is observed. All six tau isoforms
are expressed in the soluble fraction in PiD. Major tau proteins are depicted by solid bars and the
thickness of the bars correlates with the relative abundance of the specific tau isoform. A dashed
bar is used to depict the minor, and more variable, 72- and 68-kDa tau isoforms.

have also been found to be phosphorylated to some extent in tau proteins isolated
from biopsies of normal human brain (Matsuo et al 1994), it is clear that PHFtau
is hyperphosphorylated and abnormally phosphorylated (Morishima-Kawashima
et al 1995, Hasegawa et al 1996, Hoffmann et al 1997, Zheng-Fischhofer et al
1998).
P1: FUM
April 7, 2001 15:58 Annual Reviews AR121-37

NEW INSIGHTS INTO TAUOPATHIES 1129

Numerous protein kinases and protein phosphatases have been implicated in


the dysregulation of tau phosphorylation in the AD brain (for detailed reviews, see
Billingsley & Kincaid 1997, Buée et al 2000, Hong et al 2000). A recent study has
suggested that cdk5 may play a specific role in this process (Patrick et al 1999).
It showed that p25, a truncated form of p35, accumulates in neurons in the brains
of AD patients. The calcium-dependent cysteine protease calpain is believed to
generate p25 from p35 (Kusakawa et al 2000, Lee et al 2000, Nath et al 2000).
The accumulation of p25 correlated with increased cdk5 kinase activity and the
binding of p25 to cdk5 constitutively activated cdk5. Finally, the expression of the
cdk5/p25 complex in various cell lines increased tau phosphorylation and disrupted
Annu. Rev. Neurosci. 2001.24:1121-1159. Downloaded from www.annualreviews.org

the cytoskeletal network. Thus, it is possible that the cdk5/p25 complex may play
a mechanistic role in the conversion of normal tau into PHFtau in AD; however, it
will be important to confirm and extend these results in additional in vitro and in
vivo studies.
by Open University on 07/11/13. For personal use only.

The mechanisms underlying PHF formation in neurons are still unclear, but it
is possible that hyperphosphorylation disengages tau from MTs, thereby increas-
ing the pool of unbound tau. Unbound tau may be more resistant to degradation
and more prone to aggregate than MT-bound tau. This suggests that an increased
ability of pathological tau to intereact with MTs may be beneficial. The organic
osmolytes trimethylamine N-oxide and betaine have been shown to increase
tau-promoted assembly of MTs (Tseng & Graves 1998). These compounds were
also shown to restore the ability of tau phosphorylated by cAMP-dependent protein
kinase or GSK-3β to promote MT assembly (Tseng et al 1999). Tau is a highly
flexible, extended molecule with little secondary structure (Schweers et al 1994,
Goedert et al 1999a, Barghorn et al 2000). By circular dichroism spectroscopy,
it appears as a random coil (Cleveland et al 1977a), even when carrying
FTDP-17 mutations (Goedert et al 1999a, Barghorn et al 2000) or in the pres-
ence of trimethylamine N-oxide (Tseng & Graves 1998). Binding of tau to MTs
generates some ordered structures, indicating that MTs can induce conformational
changes in tau (Woody et al 1983). Trimethylamine N-oxide and betaine probably
induce a tubulin and/or tau conformational change that favors assembly.
Along related lines, Lu et al (1999) demonstrated that the prolyl isomerase Pin1
binds to a single phosphothreonine residue (Thr231) in tau and that this restored
the ability of tau phosphorylated by cdc2 kinase to intereact with MTs. Pin1 was
reported to copurify with PHFs, resulting in a depletion of soluble Pin1 in AD
brain. Because depletion of Pin1 is believed to induce mitotic arrest and apoptotic
cell death, its sequestration in NFTs could contribute to neurodegeneration.

SYNTHETIC TAU FILAMENTS

Hyperphosphorylation is believed to be an early event in the pathway that leads


from soluble to insoluble and filamentous tau protein (Braak et al 1994). However,
it is unclear whether it is sufficient for assembly into filaments. Currently, there
is no experimental evidence that links hyperphosphorylation of tau to filament
P1: FUM
April 7, 2001 15:58 Annual Reviews AR121-37

1130 LEE ¥ GOEDERT ¥ TROJANOWSKI

assembly. The availability of large quantities of recombinant human tau isoforms


and the ease with which tau fragments can be expressed has facilitated studies
aimed at producing synthetic tau filaments. Early experiments had shown that
PHF-like filaments can be assembled in vitro from bacterially expressed, non-
phosphorylated 3R fragments of tau (Crowther et al 1992, Wille et al 1992). The
formation of these filaments lent strong support to the view that the repeat region
of tau is the only component necessary for the morphological appearance of the
PHF. However, these studies failed to provide any insight into filament formation
in vivo because the tau filaments were obtained only with truncated tau under non-
physiological conditions. This contrasts with PHFs from AD brain, which consist
Annu. Rev. Neurosci. 2001.24:1121-1159. Downloaded from www.annualreviews.org

of full-length tau protein (Goedert et al 1992b).


More recently, experiments using sulphated glycosaminoglycans (GAGs) to
stimulate phosphorylation of tau by a number of protein kinases have led to the ob-
servation that sulphated GAGs induce the assembly of full-length tau into filaments
by Open University on 07/11/13. For personal use only.

(Goedert et al 1996, Perez et al 1996). Assembly of individual 3R-tau isoforms


gave filaments with a typical paired-helical-like morphology, when incubated with
heparin or heparan sulphate, whereas assembly of individual 4R-tau isoform gave
filaments with a straight appearance. By immunoelectron microscopy, the paired-
helical-like filaments were decorated by antibodies directed against the amino
and carboxy termini of tau, but not by an antibody against the MT-binding region
(Goedert et al 1996). A short amino acid sequence (VQIVYK) in the third MT-
binding repeat of tau has recently been shown to be essential for heparin-induced
filament assembly (von Bergen et al 2000).
Assembly of tau into filaments in the presence of sulphated GAGs occurs after
a lag period and is heavily concentration dependent, consistent with a nucleation-
dependent process (Goedert et al 1996, Perez et al 1996, Arrasate et al 1997,
Hasegawa et al 1997, Friedhoff et al 1998). Phosphorylation of tau at Ser/Thr-
Pro sites does not significantly influence heparin-induced assembly (Goedert et al
1996). However, it has been reported that phosphorylation at other sites, such as
Ser214 and Ser262, is strongly inhibitory toward assembly (Schneider et al 1999).
Subsequent to this work, RNA (Kampers et al 1996, Hasegawa et al 1997) and
arachidonic acid (Wilson & Binder 1997) were also shown to induce the bulk
assembly of full-length recombinant tau into filaments. This work has provided
robust methods for the assembly of full-length tau into filaments. Pathological
colocalization of sulphated GAGs (Snow et al 1990, Goedert et al 1996, Verbeek
et al 1999) and RNA (Ginsberg et al 1998) with hyperphosphorylated tau protein
suggests that these findings may also be relevant for the assembly of tau in AD.

SPORADIC TAUOPATHIES

The amyloid cascade hypothesis for the pathogenesis of AD proposes that the
deposition and fibrillization of Aβ peptides to form extracellular senile plaques is
the central event that causes formation of NFTs and neuronal loss (Hardy & Allsop
P1: FUM
April 7, 2001 15:58 Annual Reviews AR121-37

NEW INSIGHTS INTO TAUOPATHIES 1131

1991). Compelling evidence in support of a causative pathologic role of tau pro-


tein in neurodegeneration is provided by recent studies of tauopathies other than
AD, which share an abundant filamentous tau pathology and brain degeneration in
the absence of extracellular amyloid deposits (Table 1). Progressive supranuclear
palsy (PSP), corticobasal degeneration (CBD), and Pick’s disease (PiD) are three
such disorders. A recent consensus conference has classified them as disorders
belonging to a group of diseases known as frontotemporal dementia (FTD) (Pick’s
Conference 2001). Clinically, PSP is characterized by supranuclear gaze palsy as
well as by prominent postural instability (Steele et al 1964). Neuropathologically,
PSP is characterized by atrophy of the basal ganglia, subthalamus, and brainstem,
Annu. Rev. Neurosci. 2001.24:1121-1159. Downloaded from www.annualreviews.org

with corresponding neuronal loss and gliosis. Within these brain regions, there is
a high density of fibrillary tau pathology, including neuropil threads, and NFTs that
are typically round or globose (Pollock et al 1986, Hauw et al 1994, Litvan et al
1996). Glial fibrillary tangles in both astrocytes (tufted astrocytes) and oligoden-
by Open University on 07/11/13. For personal use only.

drocytes (coiled bodies) are also often present (Hauw et al 1990, Yamada et al 1992,
Komori 1999). In contrast to AD, ultrastructural analysis of these neurofibrillary
lesions has revealed 15- to 18-nm straight filaments, and filaments with a long
periodicity have also been observed (Tellez-Nagel & Wisniewski 1973, Roy et al
1974).
The filamentous tau pathology of PSP correlates with the biochemical identifi-
cation of insoluble, hyperphosphorylated tau in affected brain regions. However,
in contrast to the three major bands identified in AD, only the two high Mr bands
(68 and 64 kDa) are present (the minor 72-kDa band is variably detected) (Flament
et al 1991, Vermersch et al 1994) (Figure 2). These bands are made of hyperphos-
phorylated tau isoforms with four MT repeats (Spillantini et al 1997, Sergeant et al
1999). They exhibit the same profile of phosphorylation-dependent tau epitopes
as those detected in PHFtau from AD brains (Schmidt et al 1996). Furthermore,
in PSP, the relative abundance of tau mRNA containing E10 has been reported to
be increased in the brainstem but not in the cortex, which is consistent with the
distribution of the neurofibrillary pathology (Chambers et al 1999).
Polymorphisms in the tau gene may contribute to the risk of developing PSP
because a polymorphic dinucleotide repeat in the intron between E9 and E10 of the
tau gene has been linked to PSP (Conrad et al 1997). Subjects with the homozygous
tau allele A0, characterized by 11 TG repeats, were found to be overrepresented in
PSP patients (95.5%) compared with controls (57.4%) and AD (49.7%) patients.
Subsequent studies have confirmed this correlation in the Caucasian (but not Asian)
population (Bennett et al 1998, Higgins et al 1998, Hoenicka et al 1999, Morris
et al 1999a). Moreover, two extended tau gene haplotypes consisting of eight
common single-nucleotide polymorphisms in addition to the dinucleotide repeat
polymorphism have been described (Baker et al 1999). The haplotypes are in
complete linkage dysequilibrium and span the entire human tau gene. The more
common haplotype, H1, is significantly overrepresented in Caucasians with PSP.
In addition, two missense mutations in E4a are associated with the H1 haplotype
and have been linked to PSP, and a 238-bp deletion in the intron flanking E10 of
P1: FUM
April 7, 2001 15:58 Annual Reviews AR121-37

1132 LEE ¥ GOEDERT ¥ TROJANOWSKI

the tau gene is inherited as part of the less-common H2 extended haplotype and
thus shows a negative assciation with PSP (Higgins et al 1998, Baker et al 1999,
Bonifati et al 1999, Ezquerra et al 1999). The relationships of the H1 haplotype
and the A0 allele to the pathogenesis of PSP are unknown, but it is possible that
the 238-bp deletion flanking E10 in the H1 haplotype might affect E10 splicing,
thereby increasing the relative proportion of 4R-tau.
CBD is an adult-onset progressive neurodegenerative disorder involving the
cerebral cortex, deep cerebellar nuclei, and substantia nigra, in association with
prominent neuronal achromasia (Rebeiz et al 1967, 1968). Neuropathological ex-
amination shows depigmentation of the substantia nigra, as well as an asymmetric
Annu. Rev. Neurosci. 2001.24:1121-1159. Downloaded from www.annualreviews.org

frontoparietal atrophy that is often most severe in the pre- and postcentral regions.
In affected regions, there is neuronal loss with spongiosis, gliosis, and prominent
glial and neuronal intracytoplasmic filamentous tau pathology (Iwatsubo et al 1994,
Mori et al 1994). The glial tau pathology in CBD consists of characteristic astro-
by Open University on 07/11/13. For personal use only.

cytic plaques (Feany & Dickson 1995), as well as numerous tau-immunoreactive


inclusions in the white matter in both astrocytes and oligodendrocytes (coiled bod-
ies) (Komori et al 1998, Komori 1999). A striking feature of CBD is the extensive
accumulation of tau-immunoreactive neuropil threads throughout gray and white
matter (Feany & Dickson 1995, Feany et al 1996). The tau filaments in CBD
include both PHF-like filaments and straight tubules (Ksiezak-Reding et al 1994,
Komori 1999).
The biochemical profile of insoluble tau in CBD is similar to that of PSP in
that it consists of two major bands of 64 and 68 kDa and a variable, minor band
of 72 kDa (Ksiezak-Reding et al 1994, Buée-Scherrer et al 1996) (Figure 2).
However, the isoforms present in the tau pathology of CBD may differ from those
found in PSP. Antibodies specific for the insert encoded by E3 did not detect the
fibrillary tau pathology in CBD either biochemically or immunohistochemically in
studies from one group (Ksiezak-Reding et al 1994, Feany et al 1995). However,
another report failed to confirm this finding (Sergeant et al 1999). The latter study
demonstrated that the fibrillary inclusions in CBD are composed predominantly
of 4R-tau isoforms that also contain the inserts encoded by E2 and to E3. Another
similarity between PSP and CBD was recently described by Di Maria et al (2000),
who showed that CBD is associated with the A0 allele of the tau gene, as well as
the H1 haplotype. Thus, the current biochemical and genetic data strongly suggest
that there is a substantial overlap between PSP and CBD. This is also apparent
with respect to the clinical (Hauw et al 1994) and pathological (Feany et al 1996)
features. Rather than representing two separate and distinct disorders, PSP and
CBD may be different phenotypic manifestations of the same underlying disease
process.
PiD, a variant of FTD, is defined neuropathologically by the presence of tau-
immunoreactive Pick bodies (Constantinidis et al 1974, Pollock et al 1986, Feany
et al 1996, Pick’s Conference 2001). Neuropathologically, it is characterized
by a frontotemporal lobar and limbic atrophy associated with marked neuronal
loss, spongiosis, and gliosis, with ballooned neurons and Pick bodies (Lund &
P1: FUM
April 7, 2001 15:58 Annual Reviews AR121-37

NEW INSIGHTS INTO TAUOPATHIES 1133

Manchester Groups 1994, Dickson 1998). Pick bodies are detected by antibodies
to hyperphosphorylated tau and are most numerous in layers II and VI of the neo-
cortex and in the dentate granule neurons of the hippocampus (Iwatsubo et al 1994,
Probst et al 1996). Ultrastructurally, Pick bodies are composed of a mixture of
wide, straight filaments and wide, long-period twisted filaments (Munoz-Garcia &
Ludwin 1984, Murayama et al 1990, Dickson 1998).
Western blot analyses have revealed that the insoluble tau in PiD is distinct
from that in AD, CBD, and PSP in that it comprises two major bands of 60 and 64
kDa and a variable, minor band of 68 kDa (Buée-Scherrer et al 1996, Delacourte
et al 1996, Lieberman et al 1998) (Figure 2). Because the two major PiD tau
Annu. Rev. Neurosci. 2001.24:1121-1159. Downloaded from www.annualreviews.org

bands appear to lack the MT binding repeat encoded by E10, they are believed
to be composed exclusively of 3R-tau (Sergeant et al 1997, Mailliot et al 1998).
Ser262 has been shown not to be phosphorylated, in contrast to AD, CBD, or PSP
(Probst et al 1996, Delacourte et al 1998). However, a separate study has detected
by Open University on 07/11/13. For personal use only.

a signal in Pick bodies and PiD tau using an antibody specific for phosphorylated
Ser262 (Lieberman et al 1998). This may reflect heterogeneity of phosphorylation
at Ser262 in PiD.
In contrast to PiD, the majority of patients with FTD show frontotemporal neu-
ron loss, gliosis, and microvacuolar (spongiform) change but no disease-specific
diagnostic lesions. This neuropathological entity is referred to by several names,
including frontotemporal lobar degeneration (FTLD) and dementia lacking dis-
tinctive histology (DLDH), but there is no agreement on the most appropriate
nomenclature for this form of FTD (Mann 1998). However, this may change
soon because one study has defined this neuropathology more precisely using
quantitative morphometric methods (Arnold et al 2000). Moreover, recent evi-
dence suggests that FTLD (DLDH) may be a novel tauopathy that is caused by
a selective reduction or complete loss of all six brain tau isoforms in affected
and unaffected brain regions (Zhukareva et al 2001). The explanation for this is
enigmatic because there was no concomitant loss of tau mRNA compared with
control and AD brains. Although the majority of FTD patients with FTLD (DLDH)
neuropathology showed a dramatic loss of tau protein in brain regions with and
without neuronal degeneration, others showed less-substantial but still statistically
significant reductions in brain tau levels. Although the pathogenic mechanism un-
derlying this marked reduction in all six brain tau proteins in FTLD (DLDH) is not
known, the consequence of this loss may be similar to the losses of tau function
resulting from some of the tau gene mutations in FTDP-17.

FAMILIAL TAUOPATHIES—FTDP-17 SYNDROMES


The group of syndromes known as FTDP-17 consists of autosomal-dominantly
inherited neurodegenerative diseases with diverse, but overlapping, clinical and
neuropathological features (Foster et al 1997). Neuropathologically, they all show
the presence of an abundant filamentous tau pathology in nerve cells, and for some
P1: FUM
April 7, 2001 15:58 Annual Reviews AR121-37

1134 LEE ¥ GOEDERT ¥ TROJANOWSKI

in glial cells (reviewed in Spillantini et al 1998a, Crowther & Goedert 2000, Hong
et al 2000). The first such disorder was linked to chromosome 17 in 1994, when
Wilhelmsen et al (1994) described a familial disease they called “disinhibition-
dementia-parkinsonism-amyotrophy complex” and demonstrated genetic linkage
of this disease to chromosome 17q21-22. Subsequently, a number of related neu-
rodegenerative disorders were linked to the same region on chromosome 17 (Wijker
et al 1996, Bird et al 1997, Foster et al 1997, Heutink et al 1997, Murrell et al
1997, Lendon et al 1998). Clinically, they are characterized primarily by FTD and
parkinsonism (Foster et al 1997), but the different FTDP-17 syndromes appear to
reflect the burden of tau pathology and degeneration in brain regions known to
Annu. Rev. Neurosci. 2001.24:1121-1159. Downloaded from www.annualreviews.org

subserve specific cognitive, executive, or motor functions. Despite this pheno-


typic heterogeneity, the neuropathology of FTDP-17 is characterized by marked
neuronal loss in affected brain regions, with extensive neuronal or neuronal and
glial fibrillary pathology composed of hyperphosphorylated tau protein, but with-
by Open University on 07/11/13. For personal use only.

out evidence of Aβ deposits or other disease-specific brain lesions in the majority


of the cases (Murrell et al 1999, Lippa et al 2000, Rizzini et al 2000, Spillantini
et al 2000).
Because the tau gene had been localized to chromosome 17q21-22, it was
an obvious candidate for the disease locus. In 1998, several groups identified
pathogenic mutations in the tau gene that segregated with FTDP-17 (Clark et al
1998, Dumanchin et al 1998, Hutton et al 1998, Poorkaj et al 1998, Spillantini
et al 1998c). To date, >20 distinct pathogenic mutations in the tau gene have
been identified in a large number of families with FTDP-17 (Table 2, Figure 3).
Eleven missense mutations in coding regions of the tau gene are known, includ-
ing mutations in E9 [K257T (Pickering-Brown et al 2000, Rizzini et al 2000),
I260V (M Hutton, personal communication), and G272V (Hutton et al 1998)],
E10 [N279K (Clark et al 1998, Delisle et al 1999, Yasuda et al 1999, Arima et al
2000), P301L (Clark et al 1998, Dumanchin et al 1998, Hutton et al 1998, Bird
et al 1999, Houlden et al 1999, Mirra et al 1999, Kodama et al 2000), P301S
(Bugiani et al 1999, Sperfeld et al 1999), and S305N (Iijima et al 1999)], in E12
[V337M (Poorkaj et al 1998) and E342V (Lippa et al 2000)], and in E13 [G389R
(Murrell et al 1999, Pickering-Brown et al 2000)] and R406W (Hutton et al 1998,
Van Swieten et al 1999)]. Three silent mutations in E10 [L284L (D’Souza et al
1999), N296N (Spillantini et al 2000), and S305S (Stanford et al 2000)] as well
as a deletion mutation [1K280 (Rizzu et al 1999)] have also been identified. In
addition, five substitutions in six different positions of the intron following E10
have been identified at positions +3 (Spillantini et al 1998c, Tolnay et al 2000),
+12 (Yasuda et al 2000), +13 (Hutton et al 1998), +14 (Clark et al 1998, Hutton
et al 1998), and +16 (Hutton et al 1998, Hulette et al 1999, Goedert et al 1999b,
Morris et al 1999b). Besides mutations in the intron following E10, additional
pathogenic mutations may be present in other introns of the tau gene. Thus, a
mutation in the intron following E9 has been described in a patient with familial
FTD (Rizzu et al 1999). It disrupts one of the several (A/T)GGG repeats that may
play a role in the regulation of the alternative splicing of E10.
P1: FUM
April 7, 2001 15:58 Annual Reviews AR121-37

NEW INSIGHTS INTO TAUOPATHIES 1135

TABLE 2 Tau mutations identified in FTDP-17a


Mutation Location E10 Splicing MT Assembly Phenotype Reference

K257T E9, R1 No change Reduced PiD-like Pickering-Brown et al


(2000), Rizzini et al
(2000)
1260V E9, R1 ND ND NA M Hutton, personal
communication
G272V E9, R1 No change Reduced FTDP-17 Hutton et al (1998)
Annu. Rev. Neurosci. 2001.24:1121-1159. Downloaded from www.annualreviews.org

N279K E10, IR1-2 Increased No effect PSP-like Clark et al (1998)


1280K E10, IR1-2 Decreased Reduced FTDP-17 Rizzu et al (1999)
L284L E10, IR1-2 Increased No effect AD-like D’Souza et al (1999)
by Open University on 07/11/13. For personal use only.

N296N E10, R2 Increased No effect CBD-like Spillantini et al (2000)


P301L E10, R2 No change Reduced FTDP-17, Hutton et al (1998)
CBD-like,
PSP-like
P301S E10, R2 No change Reduced FTDP-17, Bugiani et al (1999),
CBD-like Sperfeld et al (1999)
S305N E10, IR2-3 Increased No effect CBD-like D’Souza et al (1999),
Hasegawa et al (1999),
Iijima et al (1999)
S305S E10, IR2-3 Increased No effect PSP-like Stanford et al (2000)
E10+3 I10 Increased No effect FTDP-17 Spillantini et al (1998c)
E10+12 I10 Increased No effect FTDP-17 Yasuda et al (2000)
E10+13 I10 Increased No effect NA Hutton et al (1998)
E10+14 I10 Increased No effect FTDP-17, Hutton et al (1998)
PSP-like
E10+16 I10 Increased No effect FTDP-17, Hutton et al (1998)
PSP-like
CBD-like
E9+33 I9 ND ND NA Rizzu et al (1999)
V337M E12, IR3-4 No change Reduced FTDP-17 Poorkaj et al (1998)
E342V E12, IR3-4 ND ND FTDP-17 Lippa et al (2000)
G389R E13 No change Reduced PiD-like Murrell et al (1999)
R406W E13 No change Reduced PSP-like Hutton et al (1998)
a
FTDP-17, frontotemporal demential and parkinsonism linked to chromosome 17; E, exon; I, intron; R, microtubule (MT)
binding repeat; IR, interrepeat regions; ND, not determined; NA, not available; PiD, Pick’s disease; PSP, progressive supranu-
clear palsy; AD, Alzheimer’s disease; CBD, corticobasal degeneration; increased, enhanced E10 utilization; decreased,
reduced E10 utilization.
P1: FUM
April 7, 2001 15:58 Annual Reviews AR121-37

1136 LEE ¥ GOEDERT ¥ TROJANOWSKI


Annu. Rev. Neurosci. 2001.24:1121-1159. Downloaded from www.annualreviews.org

Figure 3 Schematic representation of mutations in the tau gene identified in frontotemporal


dementia and parkinsonism linked to chromosome 17. The longest human brain tau isoform is
by Open University on 07/11/13. For personal use only.

shown with known coding region mutations indicated above. The gray boxes near the amino
terminus represent the alternatively spliced inserts encoded by exons (E)2 and E3, whereas the
black boxes represent each of the four microtubule (MT) binding repeats (not drawn to scale).
The second MT binding repeat is encoded by E10. Part of the mRNA sequence encoding E10 and
the intron following E10 is shown. Mutations in E10 and the downstream intron are indicated.
Intronic nucleotides that are part of intron 10 are shown in lower case.

Data emerging from several laboratories continue to add increasing support


in favor of the hypothesis that FTDP-17 mutations lead to tau dysfunction and
disease by one or more of three distinct mechanisms. Intronic and some exonic
mutations affect the alternative splicing of E10 and consequently alter the relative
proportion of 4R-tau and 3R-tau. The other exonic mutations impair the ability
of tau to bind MTs and to promote MT assembly. Some of these mutations also
promote the assembly of tau into filaments. Moreover, additional mechanisms
may play a role in the case of some coding region mutations (Yen et al 1999,
Goedert et al 2000). The intronic mutations clustered around the 50 splice site of
E10, as well as several mutations within E10 (N279K, L284L, N296N, S305N,
and S305S), increase the ratio of 4R-tau to 3R-tau by altering the splicing of E10
(Hong et al 1998; Hutton et al 1998; Spillantini et al 1998c; D’Souza et al 1999;
Delisle et al 1999; Grover et al 1999; Hasegawa et al 1999; Varani et al 1999;
Yasuda et al 1999, 2000; Spillantini et al 2000; Stanford et al 2000). As a result of
these mutations, there is a relative increase in E10-containing tau mRNAs, and this
probably reflects increased utilization of the E10 50 splice site, as demonstrated in
exon trapping experiments. Biochemical analysis of insoluble tau extracted from
autopsied FTDP-17 brain tissue of patients with these mutations reveals exclusively
4R-tau isoforms (Spillantini et al 1997, 1998c; Clark et al 1998; Hong et al 1998;
Reed et al 1998; Hulette et al 1999; Goedert et al 1999b; Yasuda et al 2000).
Furthermore, 4R-tau protein levels are increased in both affected and unaffected
regions of FTDP-17 brains (Hong et al 1998, Spillantini et al 1998c, Goedert et al
1999b, Yasuda et al 2000).
P1: FUM
April 7, 2001 15:58 Annual Reviews AR121-37

NEW INSIGHTS INTO TAUOPATHIES 1137

The regulation of splicing of E10 in the tau gene appears to be complex and may
involve multiple cis-acting regulatory elements that either enhance or inhibit the
utilization of the E10 50 splice site, many of which are affected by the mutations
identified in the tau gene (D’Souza et al 1999, Grover et al 1999, D’Souza &
Schellenberg 2000, Gao et al 2000, Jiang et al 2000). Splicing regulatory elements
within E10 appear to include an exon-splicing enhancer (ESE) and an exon splicing
silencer (ESS) (D’Souza et al 1999, D’Souza & Schellenberg 2000, Gao et al 2000).
The ESE consists of three domains, a potential SC35 binding element, a purine-rich
sequence, and an AC-rich sequence (D’Souza & Schellenberg 2000). Immediately
downstream of the ESE within E10 is a purine-rich ESS. The flanking exons of the
Annu. Rev. Neurosci. 2001.24:1121-1159. Downloaded from www.annualreviews.org

tau gene also appear to affect E10 splicing (Gao et al 2000). For example, it appears
that E9 and E11 exert opposite effects, i.e. E9 may promote E10 splicing, whereas
E11 may suppress it. Lastly, intronic sequences immediately downstream of E10
inhibit its splicing (D’Souza et al 1999, Grover et al 1999, D’Souza & Schellenberg
by Open University on 07/11/13. For personal use only.

2000, Gao et al 2000, Jiang et al 2000). This inhibition may be secondary to the
formation of a stem-loop structure that sequesters the E10 50 splice site from
the splicing machinery, including the U1- and U6-snRNPs (Grover et al 1999;
Varani et al 1999, 2000; Jiang et al 2000) (Figure 3). The determination of the
three-dimensional structure of a 25-nucleotide-long RNA from the E10 50-intron
junction by nuclear magnetic resonance spectroscopy has shown that this sequence
forms a stable, folded stem-loop structure (Varani et al 1999, 2000). The stem
consists of a single G-C base pair that is separated from a double helix of 6 bp by
an unpaired adenine (Figure 3). As is often the case with single-nucleotide purine
bulges, the unpaired adenine at position −2 does not extrude into solution but
intercalates into the double helix. The apical loop consists of six nucleotides that
adopt multiple conformation in rapid exchange. Known intronic mutations and
the mutations in codon 305 are located in the upper part of the stem and reduce
the thermodynamic stability of the stem loop (Varani et al 1999, 2000; Yasuda
et al 2000). Moreover, the relative proportions of 3R-tau and 4R-tau isoforms
from nonhuman species correlate with the predicted stability of this stem-loop
structure (Grover et al 1999). However, another study has concluded that this ESS
may function as a linear sequence that is independent of a stem-loop structure
(D’Souza & Schellenberg 2000).
Pathogenic FTDP-17 mutations in the tau gene may alter E10 splicing by af-
fecting several of the regulatory elements described above. Thus, the intronic
mutations, as well as the exonic mutations at codon 305 (S305N and S305S),
may destabilize the inhibitory stem-loop structure (Grover et al 1999, Varani et al
1999) (Figure 3). The S305N mutation and the +3 intronic mutation may also
enhance E10 splicing by increasing the strength of the 50 splice site (Spillantini
et al 1998c, Iijima et al 1999). However, the finding that the S305S mutation that
weakens the E10 50 splice site also leads to a predominance of 4R-tau argues against
this effect of these mutations (Stanford et al 2000). The N279K mutation may
improve the function of the ESE by lengthening the purine-rich sequence within
this regulatory element (TAAGAA to GAAGAA), thus enhancing E10 splicing
P1: FUM
April 7, 2001 15:58 Annual Reviews AR121-37

1138 LEE ¥ GOEDERT ¥ TROJANOWSKI

(D’Souza & Schellenberg 2000). Moreover, the thymidine nucleotide present in


the wild-type (WT) sequence may function as an inhibitor of splicing (Tanaka et al
1994). The observation that the 1K280 mutation, which deletes the three adjacent
purine residues (AAG), abolishes E10 splicing supports this hypothesis (D’Souza
et al 1999). The silent L284L mutation that enhances E10 splicing may do so
by disrupting a potential ESS (UUAG to UCAG) (Si et al 1998, D’Souza et al
1999). However, because mutation of this consensus sequence does not increase
E10 splicing (D’Souza & Schellenberg 2000), a second possibility is that the mu-
tation lengthens the AC-rich element within the ESE. Thus, the L284L mutation
may affect either an enhancing or an inhibiting regulatory splicing element. The
Annu. Rev. Neurosci. 2001.24:1121-1159. Downloaded from www.annualreviews.org

effect of the N296N mutation on splicing of E10 is probably due to disruption of


an ESS (D’Souza & Schellenberg 2000, Spillantini et al 2000).
The mechanisms by which these changes in the ratio of 3R-tau to 4R-tau (3R/4R-
tau) lead to neuronal and glial dysfunction and cell death remain unclear. 4R-tau
by Open University on 07/11/13. For personal use only.

and 3R-tau may bind to distinct sites on MTs (Goode & Feinstein 1994, Goode
et al 1997), and it is possible that a specific ratio of tau isoforms is necessary for
normal MT function. Thus, the altered ratio of 3R/4R-tau may directly affect MT
function. In addition, overproduction of 4R-tau may lead to an excess of free tau
in the cytoplasm, leading to its hyperphosphorylation and assembly into filaments.
In contrast to the FTDP-17 mutations discussed above, other mutations alter the
ability of tau to interact with MTs. Specifically, mutations K257T, G272V, 1K280,
P301L, P301S, V337M, G389R, and R406W reduce the binding of tau to MTs
and decrease its ability to promote MT assembly in in vitro assays (Hasegawa
et al 1998, Hong et al 1998, Bugiani et al 1999, D’Souza et al 1999, Murrell
et al 1999, Rizzu et al 1999, Barghorn et al 2000, Pickering-Brown et al 2000,
Rizzini et al 2000). These effects are not observed with the tau missense muta-
tions that affect E10 splicing (Hong et al 1998, D’Souza et al 1999, Hasegawa et al
1999). Similar effects on MT function are observed when tau is expressed in a
variety of cell lines, including SHSY5Y neuroblastoma cells (Dayanandan et al
1999), Chinese hamster ovary (CHO) cells (Dayanandan et al 1999, Matsumura
et al 1999, Vogelsberg-Ragaglia et al 2000), monkey kidney (COS) cells (Arawaka
et al 1999, Sahara et al 2000), and Sf9 insect cells (Frappier et al 1999). Expression
of a variety of tau missense mutations including G272V, 1280K, P301L, V337M,
and R406W in these cells caused varying degrees of reduced MT binding, disorga-
nized MT morphology, and defects in MT assembly and MT instability. However,
in two studies, many mutations had only a modest or no effect on MT binding
and/or function, both in in vitro assays and in transfected cell lines (DeTure et al
2000, Sahara et al 2000). The discrepancies between these and other studies are
most likely due to the differences in the levels of expression, the methods used for
the quantification of tau levels, and the binding of tau to MTs. Nevertheless, even
if these missense mutations cause only a modest reduction in MT binding affinity,
this could have large cumulative effects on affected neurons over the human life
span. Furthermore, increased cytosolic concentrations of unbound mutant tau pro-
teins may facilitate aggregation of these abnormal proteins, with or without their
WT counterparts, into filamentous inclusions.
P1: FUM
April 7, 2001 15:58 Annual Reviews AR121-37

NEW INSIGHTS INTO TAUOPATHIES 1139

A subset of missense tau gene mutations may cause FTDP-17, at least in part,
by promoting tau aggregation. Several studies have demonstrated that some of
these mutations, including K257T, G272V, 1280K, P301L, P301S, V337M, and
R406W, promote heparin- or arachidonic acid–induced tau filament formation in
vitro relative to WT tau (Arrasate et al 1999, Nacharaju et al 1999, Goedert et al
1999a, Barghorn et al 2000, Gamblin et al 2000, Rizzini et al 2000). Furthermore,
aggregation of mutant tau proteins in intact cells also has been demonstrated.
Thus, CHO cells expressing tau with the 1K280 mutation, but not other mutations
(V337M, P301L, and R406W), formed insoluble amorphous and fibrillar tau ag-
gregates (Vogelsberg-Ragaglia et al 2000). In addition, expression of the 1K280,
Annu. Rev. Neurosci. 2001.24:1121-1159. Downloaded from www.annualreviews.org

and R406W mutants in CHO and other cells led to reduced levels of tau phospho-
rylation relative to other mutant constructs and WT tau (Dayanandan et al 1999,
Matsumura et al 1999, Perez et al 2000, Sahara et al 2000, Vogelsberg-Ragaglia
et al 2000).
by Open University on 07/11/13. For personal use only.

All known mutations in the tau gene lead to the formation of filaments made
of hyperphosphorylated tau protein (Crowther & Goedert 2000). However, the
fibrillary lesions observed with mutations in E10 or the intron following E10 are
biochemically and ultrastructurally distinct from the lesions caused by mutations
that are located outside E10. Coding region mutations located outside E10 affect all
six isoforms of tau. Thus, as one might predict, tau fibrillary lesions are composed
of all six tau isoforms (Hong et al 1998, Murrell et al 1999, Van Swieten et al
1999) (Figure 2). For some mutations (V337M and R406W), the morphologies
and biochemical characteristics of tau filaments are indistinguishable from those
of AD (Spillantini et al 1996, Hong et al 1998, Van Swieten et al 1999). Other
coding region mutations located outside E10 (K252T, G272V, E342V, and G389R)
give rise to a tau pathology that closely resembles that of PiD (Spillantini et al
1998b, Murrell et al 1999, Lippa et al 2000, Rizzini et al 2000). In contrast,
mutations located within E10 itself or the intron following E10 lead to aggregation
of predominantly 4R-tau (Clark et al 1998; Hong et al 1998; Hutton et al 1998;
Reed et al 1998; Spillantini et al 1998b,c; Hulette et al 1999; Mirra et al 1999;
Nasreddine et al 1999; Goedert et al 1999b; Yasuda et al 2000). Ultrastructurally,
these lesions are composed of twisted ribbons that are similar to the filaments
observed in 4R-tau disorders, particularly CBD (Reed et al 1998, Spillantini et al
1998b, Bird et al 1999, Bugiani et al 1999, Delisle et al 1999, Hulette et al 1999,
Iijima et al 1999, Mirra et al 1999, Goedert et al 1999b, Yasuda et al 2000).
Finally, a family with a syndrome known as hereditary dysphasic disinhibition
dementia 2 (HDDD2), which appears similar to some of the syndromes seen in
FTDP-17 kindreds, has been reported to show linkage to 17q21-22 with a lod
(logarithm of odds) score of 3.68; however, no tau gene mutation or any other
genetic abnormality has been identified in this family (Lendon et al 1998). It is
surprising, however, that recent studies of three brains from affected members of
the HDDD2 kindred revealed that HDDD2 shares significant neuropathological
and biochemical abnormalities with sporadic FTD patients classified as FTLD
(DLDH) (Lendon et al 1998, Zhukareva et al 2001). As discussed above regarding
FTLD (DLDH), the loss of tau proteins in several of the HDDD2 brains, together
P1: FUM
April 7, 2001 15:58 Annual Reviews AR121-37

1140 LEE ¥ GOEDERT ¥ TROJANOWSKI

with the preservation of tau mRNA, suggests that the abundance of tau protein may
be controlled posttranscriptionally, either at the level of tau mRNA translation or
through mechanisms that regulate mRNA stability. Thus, the HDDD2 kindred
appears to be the familial counterpart of sporadic FTLD (DLDH), both of which
may define a novel and distinct tauopathy caused by a reduction of brain tau.
Although the biochemical and structural characteristics of the tau aggregates
in FTDP-17 appear to be predictable, based on our understanding of the func-
tions of tau protein and tau gene splicing, the basis for the clinical phenotypes
and topographical distributions of pathology is more enigmatic. For example, it
is not clear why the clinical and neuropathologic phenotype of individuals with
Annu. Rev. Neurosci. 2001.24:1121-1159. Downloaded from www.annualreviews.org

FTDP-17 mutations ranges from FTD (including subtypes thereof, such as PiD,
CBD, and PSP) to multisystem neurodegeneration. However, some tau gene mu-
tations cause a similar phenotype in different families or in different members of
the same family. For instance, the N279K mutation typically causes a phenotype
by Open University on 07/11/13. For personal use only.

reminiscent of PSP with superimposed dementia (Reed et al 1998, Delisle et al


1999, Yasuda et al 1999). In contrast, there are several clinical and pathologic
descriptions of families with the P301L mutation that demonstrate a highly vari-
able phenotype ranging from PSP to CBD to PiD (Spillantini et al 1998b, Bird
et al 1999, Mirra et al 1999, Nasreddine et al 1999). Even more perplexing is the
report of two brothers from one P301L family (Bird et al 1999), with frontal lobe
degeneration in one individual and PSP-like pathology in the other. Similarly, in
a family with the P301S mutation, one individual presented with FTD whereas
his son presented clinically with CBD (Bugiani et al 1999). Although only a few
reports of this kind have been published, they suggest that there is extensive over-
lap between the various tau-related disorders and that the clinical and pathologic
distinctions between them may be due to other genetic and/or epigenetic factors
that modify the effects of the primary mutation. Currently, the specific genetic
and/or environmental modifiers that might determine the phenotype of a specific
individual remain unknown, but these are fields of active investigation, and the
generation of animal models of tau-mediated neurodegeneration may facilitate
this research (Figure 4).

EXPERIMENTAL ANIMAL MODELS OF TAUOPATHIES


Experimental and TG animal models of tauopathies will serve as informative sys-
tems for elucidating the role of abnormalities in tau in the onset and progression of
a variety of neurodegenerative disorders. In addition, they may be useful models
for the development and testing of novel therapies. Early efforts to produce an-
imal models with tau pathology were largely based on the hypothesis that the
development of extracellular Aβ pathology in TG mice would induce intraneu-
ronal tau pathology. However, although various TG mouse lines accumulate Aβ
plaques, none has developed AD-like tau pathology (Games et al 1995, Hsiao et al
1996, Sturchler-Pierrat et al 1997). More recently, several animal models of tau
pathology were produced by overexpressing human tau proteins (Table 3).
P1: FUM
April 7, 2001 15:58 Annual Reviews AR121-37

NEW INSIGHTS INTO TAUOPATHIES 1141


Annu. Rev. Neurosci. 2001.24:1121-1159. Downloaded from www.annualreviews.org
by Open University on 07/11/13. For personal use only.

Figure 4 Model of disease pathways in tauopathies. Mutations and/or polymorphisms in the tau
gene in conjunction with environmental and additional genetic factors initiate pathogenic processes
that cause regional and cell type–specific tau pathology and neurodegeneration, thus leading to
specific clinicopathologic phenotypes. PiD, Pick’s disease; CBD, corticobasal degeneration; PSP,
progressive supranuclear palsy; FTDP-17, frontotemporal dementia and parkinsonism linked to
chromosome 17.

Initial reports described TG mouse lines expressing 4R2N or 3R0N human tau
utilizing cDNA constructs with either the Thy1 or the 3-hydroxy-3-methylglutaryl
coenzyme A reductase promoters (Götz et al 1995, Brion et al 1999). Both lines
developed somatodendritic expression of tau, suggestive of “pretangle” pathology,
but no filamentous tau inclusions were observed and the animals were phenotyp-
ically normal. The lack of filament formation may have been due to the rela-
tively modest expression levels of human tau, and this notion is supported by the
Annu. Rev. Neurosci. 2001.24:1121-1159. Downloaded from www.annualreviews.org
by Open University on 07/11/13. For personal use only.
P1: FUM

TABLE 3 Transgenic models of tauopathies


April 7, 2001

1142

Gene Promoter Tau Pathology Phenotype Reference

Tau, 4R/2N Thy1 Somatodendritic tau expression Normal Götz et al (1995)


LEE
15:58

Tau, 3R/0N HMG-CoA Somatodendritic tau expression Brion et al (1999)


reductase
Tau, 3R/0N Prion protein Somatodendritic tau expression; tau Axonopathy with Ishihara et al
immunoreactive spheroids in brain muscle weakness (1999, 2001)
GOEDERT

and spinal cord; neurofibrillary


¥

tangles in brain at 18 months or older


Tau, 4R/2N Thy1 Somatodendritic tau expression; tau Axonopathy with sensori- Spittaels et al
Annual Reviews

immunoreactive spheroids in brain motor dysfunction (1999)


and spinal cord
Tau, 4R/2N Thy1 Somatodendritic tau expression; tau Axonopathy with Probst et al (2000)
immunoreactive spheroids in brain muscle weakness
TROJANOWSKI

and spinal cord


AR121-37

Tau, 4R/0N, Prion protein Neurofibrillary tangles in brain and spinal Motor and behavioral Lewis et al (2000)
P301L cord; somatodendritic tau expression deficits; amyotrophy
Tau, 4R/2N, Thy1 Neurofibrillary tangles in brain and spinal Götz et al (2001)
P301L cord; somatodendritic tau expression
Tau, genomic Endogenous Axonal expression Normal Duff et al (2000)
ApoE4 Multiple Phosphorylated tau expression in Motor dysfunction Tesseur et al (2000)
neocortex, hippocampus, and amygdala and amyotrophy
p25 Neuron specific Phosphorylated tau expression in Increased loco- Ahlijanian
enolase cortex, amygdala, and thalamus motor activity et al (2000)
Anti-NGF Cytomegalovirus Phosphorylated tau expression in Spatial memory and Capsoni
IgH/Igκ early region cortex and hippocampus with object recognition et al (2000)
associated neuron loss impairment
P1: FUM
April 7, 2001 15:58 Annual Reviews AR121-37

NEW INSIGHTS INTO TAUOPATHIES 1143

finding that massive overexpression of 4R/2N human tau in lamprey reticulospinal


neurons led to the formation of PHF-like tau inclusions with degeneration of a
subset of these neurons (Hall et al 1997, 2000). Subsequently, a series of papers
has described lines of TG mice expressing high levels of 3R/0N or 4R/2N human
tau utilizing cDNA constructs with either the Thy1 or the prion protein promoter
(Ishihara et al 1999, Spittaels et al 1999, Duff et al 2000, Probst et al 2000).
These mice developed numerous abnormal tau-immunoreactive nerve cell bodies
and dendrites and large numbers of pathologically enlarged axons containing tau-
immunoreactive spheroids. These changes were most prominent in spinal cord but
were also seen in brain. They were accompanied by histological and behavioral
Annu. Rev. Neurosci. 2001.24:1121-1159. Downloaded from www.annualreviews.org

signs of amytrophy. Mice doubly transgenic for 4R2N tau and GSK-3β showed
increased levels of tau phosphorylation and a marked reduction in the number of
spheroids and associated histological and behavioral changes at 3–4 months of age
(Spittaels et al 2000). In this system, therefore, hyperphosphorylation of tau corre-
by Open University on 07/11/13. For personal use only.

lates inversely with pathology. Although the tau pathology most closely resembles
that observed in the amyotrophic lateral sclerosis/parkinsonism–dementia complex
(Matsumoto et al 1990), it differs in several respects from that found in human
diseases. Thus, besides tau, the spheroids in these TG mice also contain neurofil-
ament proteins and tubulin. They are not detected by Congo red and Thioflavin S
and do not bind Gallyas silver. However, a recent report has shown that as these
tau TG mice aged to over 18 months, modest numbers of filamentous tau tangles
with similar properties to those found in AD could be detected in hippocampus
and entorhinal cortex (Ishihara et al 2001). Moreover, like NFTs in AD, the tau
tangles in these aged mice were ubiquitinated, did not contain neurofilaments or
tubulin, and were detected by Congo red, Thioflavin S, and Gallyas silver staining
methods. It is interesting that mice transgenic for the entire human tau gene ex-
pressed all six tau isoforms but failed to develop significant pathology (Duff et al
2000).
The discovery of mutations in the tau gene in FTDP-17 is leading to the pro-
duction of TG mouse lines expressing mutant human tau in neurons and glial cells.
Lewis et al (2000) developed several lines expressing modest levels of 4R/0N tau
with and without the P301L mutation. In contrast to mice expressing WT tau, mice
expressing tau with the P301L mutation exhibited an age- and gene dose-dependent
accumulation of tau tangles in both brain and spinal cord, with associated nerve cell
loss and reactive gliosis. Similar results have been reported by Götz et al (2001)
in TG lines expressing 4R2N tau with the P301L mutation. The tau tangles found
in these mice appeared to comprise only the mutant human tau, which suggests
that the P301L mutation probably causes neurodegeneration by promoting the ag-
gregation of the mutant tau. This is supported by recent biochemical studies using
antibodies specific for P301L tau that demonstrated recovery of mutant but not
WT tau from the insoluble fraction isolated from brain tissue of individuals with
the P301L tau gene mutation (Rizzu et al 2000).
Other approaches to the development of tau pathology in TG mice have made
use of molecules known to interact with tau. For example, TG mice overexpressing
P1: FUM
April 7, 2001 15:58 Annual Reviews AR121-37

1144 LEE ¥ GOEDERT ¥ TROJANOWSKI

human p25, an activator of cdk5, develop disturbances in cytoskeletal architecture


and behavioral alterations (Ahlijanian et al 2000). However, there was no biochem-
ical evidence of an accumulation of insoluble, hyperphosphorylated tau in these
mice. TG mice expressing apolipoprotein E4, an allelic risk factor for sporadic
AD (Corder et al 1993), showed an age-dependent increase in tau phosphorylation
that correlated with the level of apolipoprotein E4 expression (Tesseur et al 2000).
Although these mice showed somatodendritic expression of phosphorylated tau,
there was no evidence of fibrillary pathology. Finally, TG mice expressing antibod-
ies to nerve growth factor inside nerve cells developed a prominent age-dependent
neurodegenerative pathology, including neuronal loss and hyperphosphorylated,
Annu. Rev. Neurosci. 2001.24:1121-1159. Downloaded from www.annualreviews.org

insoluble tau in cortex and hippocampus (Capsoni et al 2000). However, as for


the apolipoprotein E4 TG mice described above, no evidence of fibrillary tau
pathology was presented. In summary, although several TG mouse models show
features of various tau-related disorders, they still fall short of demonstrating the
by Open University on 07/11/13. For personal use only.

entire constellation of the most characteristic features of human tauopathies.

CONCLUSION

The accumulation of filamentous tau inclusions is a common feature of a wide vari-


ety of neurodegenerative disorders, many of which are distinguished by the distinct
topographic and cell type–specific distributions of inclusions. The biochemical
and ultrastructural characteristics of the tau abnormalities, which are frequently
related to the inclusion or exclusion of E10, also reveal a significant phenotypic
overlap. The discovery of multiple mutations in the tau gene that lead to the ab-
normal aggregation of tau and cause FTDP-17 demonstrates that tau dysfunction
is sufficient to produce neurodegenerative disease. The mutations lead to specific
cellular alterations, including altered expression, function, and biochemistry of tau
protein. The finding that specific polymorphisms and mutations lead to diverse
phenotypes raises the possibility that the clinical and pathological expression of
these disorders may be influenced by other genetic and epigenetic factors.
All these disorders have as a common theme accumulation of hyperphosphory-
lated tau protein in a filamentous form, which almost certainly perturbs the function
of MTs and interferes with axonal transport. It remains to be established whether
a protein kinase/phosphatase imbalance is an early mechanistic step leading to the
generation of filamentous tau in some tauopathies. Genetic and/or environmental
factors could initiate a cascade of events that leads to the abnormal phosphorylation
of tau through incompletely defined pathways (Figure 4). It also remains to be
seen whether the mere presence of tau filaments inside brain cells is sufficient to
cause them to die. Similarly, the precise mechanisms by which tau protein assem-
bles into filaments in human brain remain to be discovered. Further investigation
into the mechanisms of tau dysfunction, as well as the identification of potential
disease-modifying factors, will provide additional insight into novel strategies for
disease treatment and prevention. The development of additional animal models
P1: FUM
April 7, 2001 15:58 Annual Reviews AR121-37

NEW INSIGHTS INTO TAUOPATHIES 1145

of tauopathies that more closely recapitulate human diseases will facilitate this
undertaking.
The aggregation of tau in AD and various tauopathies is but one example of ab-
normal protein-protein interactions that result in the intracellular accumulation of
filamentous proteins. Abnormal protein aggregation is observed in a large number
of neurodegenerative disorders (Prusiner 1998, Goedert et al 1998, Trojanowski
et al 1998). Thus, besides tau pathology, AD is characterized by the extracellular
accumulation of Aβ fibrils in the form of amyloid plaques; Lewy body disorders
contain intracytoplasmic filamentous aggregates of α-synuclein; trinucleotide re-
peat disorders have intranuclear inclusions composed of fibrous polyglutamines;
Annu. Rev. Neurosci. 2001.24:1121-1159. Downloaded from www.annualreviews.org

and spongiform encephalopathies demonstrate aggregates of proteinase-resistant


prion protein. Aggregation of proteins in the brain is a common theme in a diverse
group of disorders, and insight into the pathogenesis of any one of these disorders
may have implications for our understanding of the mechanisms that underlie all
by Open University on 07/11/13. For personal use only.

these diseases.

ACKNOWLEDGMENTS
VM-YL is the John H Ware third Chair of Alzheimer’s disease research at the
University of Pennsylvania. Work done in our laboratories is supported by grants
from the National Institute of Aging of the National Institutes of Health, the Dana
Foundation, the US Alzheimer’s Association, the UK Medical Research Council,
and the UK Alzheimer’s Research Trust. We thank Drs. MS Forman and V van
Deerlin for their input.

Visit the Annual Reviews home page at www.AnnualReviews.org

LITERATURE CITED
Ahlijanian MK, Barrezueta NX, Williams RD, parkinsonism with an N279K mutation of
Jakowski A, Kowsz KP, et al. 2000. Hyper- the tau gene. Neurology 54(9):1787–95
phosphorylated tau and neurofilament and Arnold SE, Han LY, Clark CM, Grossman M,
cytoskeletal disruptions in mice overexpress- Trojanowski JQ. 2000. Quantitative neuro-
ing human p25, an activator of cdk5. Proc. histological features of frontotemporal de-
Natl. Acad. Sci. USA 97(6):2910–15 generation. Neurobiol. Aging 21(6):913–919
Andreadis A, Brown WM, Kosik KS. 1992. Arrasate M, Perez M, Armas-Portela R, Avila J.
Structure and novel exons of the human tau 1999. Polymerization of tau peptides into fib-
gene. Biochemistry 31(43):10626–33 rillar structures. The effect of FTDP-17 mu-
Arawaka S, Usami M, Sahara N, Schellenberg tations. FEBS Lett. 446(1):199–202
GD, Lee G, Mori H. 1999. The tau mutation Arrasate M, Perez M, Valpuesta JM, Avila J.
(val337met) disrupts cytoskeletal networks 1997. Role of glycosaminoglycans in de-
of microtubules. NeuroReport 10(5):993–97 termining the helicity of paired helical fil-
Arima K, Kowalska A, Hasegawa M, aments. Am. J. Pathol. 151(4):1115–22
Mukoyama M, Watanabe R, et al. 2000. Two Baker M, Litvan I, Houlden H, Adamson J,
brothers with frontotemporal dementia and Dickson D, et al. 1999. Association of an
P1: FUM
April 7, 2001 15:58 Annual Reviews AR121-37

1146 LEE ¥ GOEDERT ¥ TROJANOWSKI

extended haplotype in the tau gene with three nonAlzheimer families and kindreds
progressive supranuclear palsy. Hum. Mol. with late-onset FAD. Neurology 48(4):949–
Genet. 8(4):711–15 54
Barghorn S, Zheng-Fischhöfer Q, Ackmann M, Bonifati V, Joosse M, Nicholl DJ, Vanacore
Biernat J, von Bergen M, et al. 2000. Struc- N, Bennett P, et al. 1999. The tau gene in
ture, microtubule interactions, and paired he- progressive supranuclear palsy: exclusion of
lical filament aggregation by tau mutants mutations in coding exons and exon 10 splice
of frontotemporal dementias. Biochemistry sites, and identification of a new intronic vari-
39(38):11714–21 ant of the disease-associated H1 haplotype
Baudier J, Cole RD. 1987. Phosphorylation of in Italian cases. Neurosci. Lett. 274(1):61–
tau proteins to a state like that in Alzheimer’s 65
Annu. Rev. Neurosci. 2001.24:1121-1159. Downloaded from www.annualreviews.org

brain is catalyzed by a calcium/calmodulin- Braak E, Braak H, Mandelkow EM. 1994. A se-


dependent kinase and modulated by phos- quence of cytoskeleton changes related to the
pholipids. J. Biol. Chem. 262(36):17577–83 formation of neurofibrillary tangles and neu-
Baumann K, Mandelkow EM, Biernat J, ropil threads. Acta Neuropathol. 87(6):554–
Piwnica-Worms H, Mandelkow E. 1993. 67
by Open University on 07/11/13. For personal use only.

Abnormal Alzheimer-like phosphorylation Bramblett GT, Goedert M, Jakes R, Merrick SE,


of tau-protein by cyclin-dependent kinases Trojanowski JQ, Lee VM-Y. 1993. Abnormal
cdk2 and cdk5. FEBS Lett. 336(3):417–24 tau phosphorylation at Ser396 in Alzheimer’s
Bennett P, Bonifati V, Bonuccelli U, Colosimo disease recapitulates development and con-
C, De Mari M, et al. 1998. Direct genetic ev- tributes to reduced microtubule binding. Neu-
idence for involvement of tau in progressive ron 10(6):1089–99
supranuclear palsy. European Study Group Brion JP, Passareiro H, Nunez J, Flament-
on Atypical Parkinsonism Consortium. Neu- Durand J. 1985. Mise en évidence im-
rology 51(4):982–85 munologique de la protéine tau au niveau des
Biernat J, Gustke N, Drewes G, Mandelkow lésions de dégénérescence neurofibrillaire de
EM, Mandelkow E. 1993. Phosphorylation la maladie d’Alzheimer. Arch. Biol. 95:229–
of Ser262 strongly reduces binding of tau to 35
microtubules: distinction between PHF-like Brion JP, Tremp G, Octave JN. 1999. Trans-
immunoreactivity and microtubule binding. genic expression of the shortest human tau
Neuron 11(1):153–63 affects its compartmentalization and its phos-
Billingsley ML, Kincaid RL. 1997. Regulated phorylation as in the pretangle stage of
phosphorylation and dephosphorylation of Alzheimer’s disease. Am. J. Pathol. 154(1):
tau protein: effects on microtubule interac- 255–70
tion, intracellular trafficking and neurode- Buée L, Bussière T, Buée-Scherrer V, Dela-
generation. Biochem. J. 323(3):577–91 courte A, Hof PR. 2000. Tau protein iso-
Binder LI, Frankfurter A, Rebhun LI. 1985. The forms, phosphorylation and role in neu-
distribution of tau in the mammalian central rodegenerative disorders. Brain Res. Rev.
nervous system. J. Cell Biol. 101(4):1371–78 33:(1):95–130
Bird TD, Nochlin D, Poorkaj P, Cherrier M, Buée-Scherrer V, Hof PR, Buée L, Leveugle B,
Kaye J, et al. 1999. A clinical pathological Vermersch P, et al. 1996. Hyperphosphory-
comparison of three families with frontotem- lated tau proteins differentiate corticobasal
poral dementia and identical mutations in the degeneration and Pick’s disease. Acta Neu-
tau gene (P301L). Brain 122(4):741–56 ropathol. 91(4):351–59
Bird TD, Wijsman EM, Nochlin D, Leehey Bugiani O, Murrell JR, Giaccone G, Hasegawa
M, Sumi SM, et al. 1997. Chromosome 17 M, Ghigo G, et al. 1999. Frontotemporal de-
and hereditary dementia: linkage studies in mentia and corticobasal degeneration in a
P1: FUM
April 7, 2001 15:58 Annual Reviews AR121-37

NEW INSIGHTS INTO TAUOPATHIES 1147

family with a P301S mutation in tau. J. Neu- dose of apolipoprotein E type 4 allele and the
ropathol. Exp. Neurol. 58(6):667–77 risk of Alzheimer’s disease in late onset fam-
Butner KA, Kirschner MW. 1991. Tau protein ilies. Science 261(5123):921–23
binds to microtubules through a flexible ar- Couchie D, Mavilia C, Georgieff IS, Liem RK,
ray of distributed weak sites. J. Cell Biol. Shelanski ML, Nunez J. 1992. Primary struc-
115(3):717–30 ture of high molecular weight tau present in
Capsoni S, Ugolini G, Comparini A, Ruberti F, the peripheral nervous system. Proc. Natl.
Berardi N, Cattaneo A. 2000. Alzheimer-like Acad. Sci. USA 89(10):4378–81
neurodegeneration in aged antinerve growth Crowther RA. 1991. Straight and paired helical
factor transgenic mice. Proc. Natl. Acad. Sci. filaments in Alzheimer disease have a com-
USA 97(12):6826–31 mon structural unit. Proc. Natl. Acad. Sci.
Annu. Rev. Neurosci. 2001.24:1121-1159. Downloaded from www.annualreviews.org

Chambers CB, Lee JM, Troncoso JC, Reich S, USA 88(6):2288–92


Muma NA. 1999. Overexpression of 4R-tau Crowther RA, Goedert M. 2000. Abnormal
mRNA isoforms in progressive supranuclear tau-containing filaments in neurodegenera-
palsy but not in Alzheimer’s disease. Ann. tive diseases. J. Struct. Biol. 130(2–3):271–
Neurol. 46(3):325–32 79
by Open University on 07/11/13. For personal use only.

Clark LN, Poorkaj P, Wszolek Z, Geschwind Crowther RA, Olesen OF, Jakes R, Goedert
DH, Nasreddine ZS, et al. 1998. Pathogenic M. 1992. The microtubule binding repeats
implications of mutations in the tau gene of tau protein assemble into filaments like
in pallido-ponto-nigral degeneration and re- those found in Alzheimer’s disease. FEBS
lated neurodegenerative disorders linked to Lett. 309(2):199–202
chromosome 17. Proc. Natl. Acad. Sci. USA Crowther RA, Wischik CM. 1985. Image re-
95(22):13103–7 construction of the Alzheimer paired helical
Cleveland DW, Hwo SY, Kirschner MW. 1977a. filament. EMBO J. 4(13B):3661–65
Physical and chemical properties of purified Dayanandan R, van Slegtenhorst M, Mack TG,
tau factor and the role of tau in microtubule Ko L, Yen SH, et al. 1999. Mutations in tau
assembly. J. Mol. Biol. 116(2):227–47 reduce its microtubule binding properties in
Cleveland DW, Hwo SY, Kirschner MW. 1977b. intact cells and affect its phosphorylation.
Purification of tau, a microtubule-associated FEBS Lett. 446(2–3):228–32
protein that induces assembly of micro- Delacourte A, Robitaille Y, Sergeant N, Buée L,
tubules from purified tubulin. J. Mol. Biol. Hof PR, et al. 1996. Specific pathological Tau
116(2):207–25 protein variants characterize Pick’s disease.
Cohen P. 1999. The Croonian Lecture 1998. J. Neuropathol. Exp. Neurol. 55(2):159–68
Identification of a protein kinase cascade of Delacourte A, Sergeant N, Wattez A, Gauvreau
major importance in insulin signal transduc- D, Robitaille Y. 1998. Vulnerable neuronal
tion. Philos. Trans. R. Soc. London Ser. B subsets in Alzheimer’s and Pick’s disease
354:485–95 are distinguished by their tau isoform dis-
Conrad C, Andreadis A, Trojanowski JQ, Dick- tribution and phosphorylation. Ann. Neurol.
son DW, Kang D, et al. 1997. Genetic ev- 43(2):193–204
idence for the involvement of tau in pro- Delisle MB, Murrell JR, Richardson R, Tro-
gressive supranuclear palsy. Ann. Neurol. fatter JA, Rascol O, et al. 1999. A muta-
41(2):277–81 tion at codon 279 (N279K) in exon 10 of
Constantinidis J, Richard J, Tissot R. 1974. the Tau gene causes a tauopathy with demen-
Pick’s disease. Histological and clinical cor- tia and supranuclear palsy. Acta Neuropathol.
relations. Eur. Neurol. 11(4):208–17 98(1):62–77
Corder EH, Saunders AM, Strittmatter WJ, DeTure M, Ko LW, Yen S, Nacharaju P, Eas-
Schmechel DE, Gaskell PC, et al. 1993. Gene son C, et al. 2000. Missense tau mutations
P1: FUM
April 7, 2001 15:58 Annual Reviews AR121-37

1148 LEE ¥ GOEDERT ¥ TROJANOWSKI

identified in FTDP-17 have a small effect nomic and cDNA tau transgenes. Neurobiol.
on tau-microtubule interactions. Brain Res. Dis. 7(2):87–98
853(1):5–14 Dumanchin C, Camuzat A, Campion D, Verpil-
Dickson DW. 1998. Pick’s disease: a modern lat P, Hannequin D, et al. 1998. Segregation
approach. Brain Pathol. 8(2):339–54 of a missense mutation in the microtubule-
Di Maria E, Tabaton M, Vigo T, Abbruzzese associated protein tau gene with familial
G, Bellone E, et al. 2000. Corticobasal de- frontotemporal dementia and parkinsonism.
generation shares a common genetic back- Hum. Mol. Genet. 7(11):1825–29
ground with progressive supranuclear palsy. Ezquerra M, Pastor P, Valldeoriola F, Molin-
Ann. Neurol. 47(3):374–77 uevo JL, Blesa R, et al. 1999. Identification of
Drechsel DN, Hyman AA, Cobb MH, a novel polymorphism in the promoter region
Annu. Rev. Neurosci. 2001.24:1121-1159. Downloaded from www.annualreviews.org

Kirschner MW. 1992. Modulation of the of the tau gene highly associated to progres-
dynamic instability of tubulin assembly by sive supranuclear palsy in humans. Neurosci.
the microtubule-associated protein tau. Mol. Lett. 275(3):183–86
Biol. Cell 3(10):1141–54 Feany MB, Dickson DW. 1995. Widespread
Drewes G, Ebneth A, Preuss U, Mandelkow cytoskeletal pathology characterizes cor-
by Open University on 07/11/13. For personal use only.

EM, Mandelkow E. 1997. MARK, a novel ticobasal degeneration. Am. J. Pathol.


family of protein kinases that phosphory- 146(6):1388–96
late microtubule-associated proteins and trig- Feany MB, Ksiezak-Reding H, Liu WK, Vin-
ger microtubule disruption. Cell 89(2):297– cent I, Yen SH, Dickson DW. 1995. Epi-
308 tope expression and hyperphosphorylation of
Drewes G, Lichtenberg-Kraag B, Döring F, tau protein in corticobasal degeneration: dif-
Mandelkow EM, Biernat J, et al. 1992. Mi- ferentiation from progressive supranuclear
togen activated protein (MAP) kinase trans- palsy. Acta Neuropathol. 90(1):37–43
forms tau protein into an Alzheimer-like Feany MB, Mattiace LA, Dickson DW.
state. EMBO J. 11(6):2131–38 1996. Neuropathologic overlap of progres-
Drewes G, Mandelkow EM, Baumann K, sive supranuclear palsy, Pick’s disease and
Goris J, Merlevede W, Mandelkow E. corticobasal degeneration. J. Neuropathol.
1993. Dephosphorylation of tau protein and Exp. Neurol. 55(1):53–67
Alzheimer paired helical filaments by cal- Flament S, Delacourte A, Verny M, Hauw JJ,
cineurin and phosphatase-2A. FEBS Lett. Javoy-Agid F. 1991. Abnormal tau proteins
336(3):425–32 in progressive supranuclear palsy. Similari-
D’Souza I, Poorkaj P, Hong M, Nochlin D, Lee ties and differences with the neurofibrillary
VM-Y, et al. 1999. Missense and silent tau degeneration of the Alzheimer type. Acta
gene mutations cause frontotemporal demen- Neuropathol. 81(6):591–96
tia with parkinsonism-chromosome 17 type, Foster NL, Wilhelmsen K, Sima AA, Jones MZ,
by affecting multiple alternative RNA splic- D’Amato CJ, et al. 1997. Frontotemporal de-
ing regulatory elements. Proc. Natl. Acad. mentia and parkinsonism linked to chromo-
Sci. USA 96(10):5598–603 some 17: a consensus conference. Ann. Neu-
D’Souza I, Schellenberg D. 2000. Determinants rol. 41(6):706–15
of 4 repeat tau expression: coordination be- Frappier T, Liang NS, Brown K, Leung CL,
tween enhancing and inhibitory splicing se- Lynch T, et al. 1999. Abnormal microtubule
quences for exon 10 inclusion. J. Biol. Chem. packing in processes of SF9 cells express-
275(23):17700–9 ing the FTDP-17 V337M tau mutation. FEBS
Duff K, Knight H, Refolo LM, Sanders S, Yu X, Lett. 455(3):262–66
et al. 2000. Characterization of pathology in Friedhoff P, von Bergen M, Mandelkow EM,
transgenic mice over-expressing human ge- Davies P, Mandelkow E. 1998. A nucleated
P1: FUM
April 7, 2001 15:58 Annual Reviews AR121-37

NEW INSIGHTS INTO TAUOPATHIES 1149

assembly mechanism of Alzheimer paired mutations on heparin-induced assembly of


helical filaments. Proc. Natl. Acad. Sci. USA tau filaments. FEBS Lett. 450(3):306–11
95(26):15712–17 Goedert M, Jakes R, Crowther RA, Six J, Lübke
Gamblin TC, King ME, Dawson H, Vitek MP, U, et al. 1993. The abnormal phosphoryla-
Kuret J, et al. 2000. In vitro polymeriza- tion of tau protein at Ser202 in Alzheimer
tion of tau protein monitored by laser light disease recapitulates phosphorylation during
scattering: method and application to the development. Proc. Natl. Acad. Sci. USA
study of FTDP-17 mutants. Biochemistry 90(11):5066–70
39(20):6136–44 Goedert M, Jakes R, Qi Z, Wang JH, Cohen P.
Games D, Adams D, Alessandrini R, Barbour 1995a. Protein phosphatase 2A is the major
R, Berthelette P, et al. 1995. Alzheimer-type enzyme in brain that dephosphorylates tau
Annu. Rev. Neurosci. 2001.24:1121-1159. Downloaded from www.annualreviews.org

neuropathology in transgenic mice overex- protein phosphorylated by proline-directed


pressing V717F beta-amyloid precursor pro- protein kinases or cyclic AMP-dependent
tein. Nature 373(6514):523–27 protein kinase. J. Neurochem. 65(6):2804–
Gao QS, Memmott J, Lafyatis R, Stamm S, 7
Sreaton G, Andreadis A. 2000. Complex Goedert M, Jakes R, Spillantini MG, Hasegawa
by Open University on 07/11/13. For personal use only.

regulation of tau exon 10, whose missplic- M, Smith MJ, Crowther RA. 1996. As-
ing causes frontotemporal dementia. J. Neu- sembly of microtubule-associated protein
rochem. 74(2):490–500 tau into Alzheimer-like filaments induced
Georgieff IS, Liem RK, Mellado W, Nunez J, by sulphated glycosaminoglycans. Nature
Shelanski ML. 1991. High molecular weight 383(6600):550–53
tau: preferential localization in the periph- Goedert M, Satumtira S, Jakes R, Smith MJ,
eral nervous system. J. Cell Sci. 100(1):55– Kamibayashi C, et al. 2000. Reduced binding
60 of protein phosphatase 2A to tau protein with
Ginsberg SD, Galvin JE, Chiu TS, Lee VM- frontotemporal dementia and parkinsonism
Y, Masliah E, Trojanowski JQ. 1998. RNA linked to chromosome 17 mutations. J. Neu-
sequestration to pathological lesions of neu- rochem. 75(5):2155–62
rodegenerative diseases. Acta Neuropathol. Goedert M, Spillantini MG, Cairns NJ,
96(5):487–94 Crowther RA. 1992b. Tau proteins of
Goedert M, Cohen ES, Jakes R, Cohen P. 1992a. Alzheimer paired helical filaments: abnor-
p42 MAP kinase phosphorylation sites in mal phosphorylation of all six brain isoforms.
microtubule-associated protein tau are de- Neuron 8(1):159–68
phosphorylated by protein phosphatase 2A1. Goedert M, Spillantini MG, Crowther RA.
Implications for Alzheimer’s disease. FEBS 1992c. Cloning of a big tau microtubule-
Lett. 312(1):95–99 associated protein characteristic of the pe-
Goedert M, Hasegawa M, Jakes R, Lawler ripheral nervous system. Proc. Natl. Acad.
S, Cuenda A, Cohen P. 1997. Phosphoryla- Sci. USA 89(5):1983–87
tion of microtubule-associated protein tau by Goedert M, Spillantini MG, Crowther RA,
stress-activated protein kinases. FEBS Lett. Chen SG, Parchi P, et al. 1999b. Tau gene
409(1):57–62 mutation in familial progressive subcortical
Goedert M, Jakes R. 1990. Expression of sep- gliosis. Nat. Med. 5(4):454–57
arate isoforms of human tau protein: corre- Goedert M, Spillantini MG, Davies SW. 1998.
lation with the tau pattern in brain and ef- Filamentous nerve cell inclusions in neurode-
fects on tubulin polymerization. EMBO J. generative diseases. Curr. Opin. Neurobiol.
9(13):4225–30 8(5):619–32
Goedert M, Jakes R, Crowther RA. 1999a. Ef- Goedert M, Spillantini MG, Jakes R, Crowther
fects of frontotemporal dementia FTDP-17 RA, Vanmechelen E, et al. 1995b. Molecular
P1: FUM
April 7, 2001 15:58 Annual Reviews AR121-37

1150 LEE ¥ GOEDERT ¥ TROJANOWSKI

dissection of the paired helical filament. Neu- Greenberg SG, Davies P, Schein JD, Binder LI.
robiol. Aging 16(3):325–34 1992. Hydrofluoric acid-treated tau PHF pro-
Goedert M, Spillantini MG, Jakes R, Ruther- teins display the same biochemical properties
ford D, Crowther RA. 1989a. Multiple as normal tau. J. Biol. Chem. 267(1):564–69
isoforms of human microtubule-associated Grover A, Houlden H, Baker M, Adamson J,
protein tau: sequences and localization in Lewis J, et al. 1999. 50 splice site mutations
neurofibrillary tangles of Alzheimer’s dis- in tau associated with the inherited demen-
ease. Neuron 3(4):519–26 tia FTDP-17 affect a stem-loop structure that
Goedert M, Spillantini MG, Potier MC, Ul- regulates alternative splicing of exon 10. J.
rich J, Crowther RA. 1989b. Cloning and Biol. Chem. 274(21):15134–43
sequencing of the cDNA encoding an iso- Grundke-Iqbal I, Iqbal K, Tung YC, Quinlan
Annu. Rev. Neurosci. 2001.24:1121-1159. Downloaded from www.annualreviews.org

form of microtubule-associated protein tau M, Wisniewski HM, Binder LI. 1986. Ab-
containing four tandem repeats: differential normal phosphorylation of the microtubule-
expression of tau protein mRNAs in human associated protein tau (tau) in Alzheimer cy-
brain. EMBO J. 8(2):393–99 toskeletal pathology. Proc. Natl. Acad. Sci.
Goedert M, Wischik CM, Crowther RA, Walker USA 83(13):4913–17
by Open University on 07/11/13. For personal use only.

JE, Klug A. 1988. Cloning and sequenc- Gustke N, Trinczek B, Biernat J, Mandelkow
ing of the cDNA encoding a core protein EM, Mandelkow E. 1994. Domains of tau
of the paired helical filament of Alzheimer protein and interactions with microtubules.
disease: identification as the microtubule- Biochemistry 33(32):9511–22
associated protein tau. Proc. Natl. Acad. Sci. Hall GF, Chu B, Lee G, Yao J. 2000. Human
USA 85(11):4051–55 tau filaments induce microtubule and synapse
Goode BL, Denis PE, Panda D, Radeke MJ, loss in an in vivo model of neurofibrillary de-
Miller HP, et al. 1997. Functional interactions generative disease. J. Cell Sci. 113(8):1373–
between the proline-rich and repeat regions 87
of tau enhance microtubule binding and as- Hall GF, Yao J, Lee G. 1997. Human tau be-
sembly. Mol. Biol. Cell 8(2):353–65 comes phosphorylated and forms filamen-
Goode BL, Feinstein SC. 1994. Identification tous deposits when overexpressed in lamprey
of a novel microtubule binding and assem- central neurons in situ. Proc. Natl. Acad. Sci.
bly domain in the developmentally regu- USA 94(9):4733–38
lated interrepeat region of tau. J. Cell Biol. Hanger DP, Hughes K, Woodgett JR, Brion
124(5):769–82 JP, Anderton BH. 1992. Glycogen syn-
Götz J, Chen F, Barmettler R, Nitsch RM. thase kinase-3 induces Alzheimer’s disease-
2001. Tau filament formation in transgenic like phosphorylation of tau: generation of
mice expressing P301L tau. J. Biol. Chem. paired helical filament epitopes and neuronal
276(1):529–34 localisation of the kinase. Neurosci. Lett.
Götz J, Probst A, Spillantini MG, Schafer T, 147(1):58–62
Jakes R, et al. 1995. Somatodendritic lo- Harada A, Oguchi K, Okabe S, Kuno J, Terada
calization and hyperphosphorylation of tau S, et al. 1994. Altered microtubule organiza-
protein in transgenic mice expressing the tion in small-calibre axons of mice lacking
longest human brain tau isoform. EMBO J. tau protein. Nature 369(6480):488–91
14(7):1304–13 Hardy J, Allsop D. 1991. Amyloid deposi-
Greenberg SG, Davies P. 1990. A preparation of tion as the central event in the aetiology of
Alzheimer paired helical filaments that dis- Alzheimer’s disease. Trends Pharmacol. Sci.
plays distinct tau proteins by polyacrylamide 12(10):383–88
gel electrophoresis. Proc. Natl. Acad. Sci. Hasegawa M, Crowther RA, Jakes R, Goed-
USA 87(15):5827–31 ert M. 1997. Alzheimer-like changes in
P1: FUM
April 7, 2001 15:58 Annual Reviews AR121-37

NEW INSIGHTS INTO TAUOPATHIES 1151

microtubule-associated protein tau induced and not the alpha-synuclein gene. Neurology
by sulfated glycosaminoglycans. Inhibition 50(1):270–73
of microtubule binding, stimulation of phos- Himmler A, Drechsel D, Kirschner MW, Martin
phorylation, and filament assembly depend DW Jr. 1989. Tau consists of a set of proteins
on the degree of sulfation. J. Biol. Chem. with repeated C-terminal microtubule-
272(52):33118–24 binding domains and variable N-terminal
Hasegawa M, Jakes R, Crowther RA, Lee domains. Mol. Cell Biol. 9(4):1381–
VM-Y, Ihara Y, Goedert M. 1996. Char- 88
acterization of mAb AP422, a novel Hoenicka J, Perez M, Perez-Tur J, Barabash A,
phosphorylation-dependent monoclonal an- Godoy M, et al. 1999. The tau gene A0 allele
tibody against tau protein. FEBS Lett. and progressive supranuclear palsy. Neurol-
Annu. Rev. Neurosci. 2001.24:1121-1159. Downloaded from www.annualreviews.org

384(1):25–30 ogy 53(6):1219–25


Hasegawa M, Smith MJ, Goedert M. 1998. Tau Hoffmann R, Lee VM-Y, Leight S, Varga I,
proteins with FTDP-17 mutations have a re- Otvos L Jr. 1997. Unique Alzheimer’s dis-
duced ability to promote microtubule assem- ease paired helical filament specific epitopes
bly. FEBS Lett. 437(3):207–10 involve double phosphorylation at specific
by Open University on 07/11/13. For personal use only.

Hasegawa M, Smith MJ, Iijima M, Tabira sites. Biochemistry 36(26):8114–24


T, Goedert M. 1999. FTDP-17 mutations Hong M, Chen DC, Klein PS, Lee VM-Y. 1997.
N279K and S305N in tau produce increased Lithium reduces tau phosphorylation by inhi-
splicing of exon 10. FEBS Lett. 443(2):93–96 bition of glycogen synthase kinase-3. J. Biol.
Hauw JJ, Daniel SE, Dickson D, Horou- Chem. 272(40):25326–32
pian DS, Jellinger K, et al. 1994. Pre- Hong M, Lee VM-Y. 1997. Insulin and insulin-
liminary NINDS neuropathologic criteria like growth factor-1 regulate tau phospho-
for Steele-Richardson-Olszewski syndrome rylation in cultured human neurons. J. Biol.
(progressive supranuclear palsy). Neurology Chem. 272(31):19547–53
44(11):2015–19 Hong M, Trojanowski JQ, Lee VM-Y. 2000.
Hauw JJ, Verny M, Delaere P, Cervera P, He Tau-based neurofibrillary lesions. In Neu-
Y, Duyckaerts C. 1990. Constant neurofib- rodegenerative Dementias, ed. CM Clark, JQ
rillary changes in the neocortex in progres- Trojanowski, pp. 161–75. New York: Mc-
sive supranuclear palsy. Basic differences Graw Hill
with Alzheimer’s disease and aging. Neu- Hong M, Zhukareva V, Vogelsberg-Ragaglia V,
rosci. Lett. 119(2):182–86 Wszolek Z, Reed L, et al. 1998. Mutation-
Heutink P, Stevens M, Rizzu P, Bakker E, specific functional impairments in distinct
Kros JM, et al. 1997. Hereditary frontotem- tau isoforms of hereditary FTDP-17. Science
poral dementia is linked to chromosome 282(5395):1914–17
17q21-q22: a genetic and clinicopatholog- Houlden H, Baker M, Adamson J, Grover A,
ical study of three Dutch families. Ann. Neu- Waring S, et al. 1999. Frequency of tau
rol. 41(2):150–59 mutations in three series of non-Alzheimer’s
Hiesberger T, Trommsdorff M, Howell BW, degenerative dementia. Ann. Neurol. 46(2):
Goffinet A, Mumby MC, et al. 1999. Di- 243–48
rect binding of Reelin to VLDL receptor and Hsiao K, Chapman P, Nilsen S, Eckman C,
ApoE receptor 2 induces tyrosine phosphory- Harigaya Y, et al. 1996. Correlative memory
lation of disabled-1 and modulates tau phos- deficits, Aβ elevation, and amyloid plaques
phorylation. Neuron 24(2):481–89 in transgenic mice. Science 274(5284):99–
Higgins JJ, Litvan I, Pho LT, Li W, Nee LE. 102
1998. Progressive supranuclear gaze palsy Hulette CM, Pericak-Vance MA, Roses AD,
is in linkage disequilibrium with the tau Schmechel DE, Yamaoka LH, et al. 1999.
P1: FUM
April 7, 2001 15:58 Annual Reviews AR121-37

1152 LEE ¥ GOEDERT ¥ TROJANOWSKI

Neuropathological features of frontotempo- aggregation of microtubule-associated pro-


ral dementia and parkinsonism linked to tein tau into Alzheimer-like paired helical fil-
chromosome 17q21–22 (FTDP-17): Duke aments. FEBS Lett. 399(3):344–49
Family 1684. J. Neuropathol. Exp. Neurol. Kanemaru K, Takio K, Miura R, Titani K, Ihara
58(8):859–66 Y. 1992. Fetal-type phosphorylation of the
Hutton M, Lendon CL, Rizzu P, Baker M, tau in paired helical filaments. J. Neurochem.
Froelich S, et al. 1998. Association of mis- 58(5):1667–75
sense and 50 -splice-site mutations in tau Kidd M. 1963. Paired helical filaments in elec-
with the inherited dementia FTDP-17. Na- tron microscopy of Alzheimer’s disease. Na-
ture 393(6686):702–5 ture 197:192–94
Iijima M, Tabira T, Poorkaj P, Schellenberg GD, Kobayashi S, Ishiguro K, Omori A, Takamatsu
Annu. Rev. Neurosci. 2001.24:1121-1159. Downloaded from www.annualreviews.org

Trojanowski JQ, et al. 1999. A distinct fa- M, Arioka M, et al. 1993. A cdc2-related
milial presenile dementia with a novel mis- kinase PSSALRE/cdk5 is homologous with
sense mutation in the tau gene. NeuroReport the 30 kDa subunit of tau protein kinase II,
10(3):497–501 a proline-directed protein kinase associated
Ikegami S, Harada A, Hirokawa N. 2000. Mus- with microtubule. FEBS Lett. 335(2):171–75
by Open University on 07/11/13. For personal use only.

cle weakness, hyperactivity, and impairment Kodama K, Okada S, Iseki E, Kowalska A,


in fear conditioning in tau-deficient mice. Tabira T, et al. 2000. Familial frontotempo-
Neurosci. Lett. 279(3):129–32 ral dementia with a P301L tau mutation in
Ishiguro K, Kobayashi S, Omori A, Takamatsu Japan. J. Neurol. Sci. 176(1):57–64
M, Yonekura S, et al. 1994. Identification of Komori T. 1999. Tau-positive glial inclusions
the 23 kDa subunit of tau protein kinase II as in progressive supranuclear palsy, corti-
a putative activator of cdk5 in bovine brain. cobasal degeneration and Pick’s disease.
FEBS Lett. 342(2):203–8 Brain Pathol. 9(4):663–79
Ishihara T, Hong M, Zhang B, Nakagawa Y, Lee Komori T, Arai N, Oda M, Nakayama H, Mori
MK, et al. 1999. Age-dependent emergence H, et al. 1998. Astrocytic plaques and tufts of
and progression of a tauopathy in transgenic abnormal fibers do not coexist in corticobasal
mice overexpressing the shortest human tau degeneration and progressive supranuclear
isoform. Neuron 24(3):751–62 palsy. Acta Neuropathol. 96(4):401–8
Ishihara T, Zhang B, Higuchi M, Yashiyama Y, Kondo J, Honda T, Mori H, Hamada Y, Miura
Trojanowski JQ, Lee VM-Y. 2001. Age de- R, et al. 1988. The carboxyl third of tau
pendent induction of congophilic neurofib- is tightly bound to paired helical filaments.
rillary inclusions in tau transgenic mice. Am. Neuron 1(9):827–34
J. Pathol. 158(2):555–62 Kosik KS, Orecchio LD, Binder L, Trojanowski
Iwatsubo T, Hasegawa M, Ihara Y. 1994. Neu- JQ, Lee VM-Y, Lee G. 1988. Epitopes that
ronal and glial tau-positive inclusions in span the tau molecule are shared with paired
diverse neurologic diseases share common helical filaments. Neuron 1(9):817–25
phosphorylation characteristics. Acta Neu- Ksiezak-Reding H, Morgan K, Mattiace LA,
ropathol. 88(2):129–36 Davies P, Liu WK, et al. 1994. Ultrastructure
Jiang Z, Cote J, Kwon JM, Goate AM, Wu JY. and biochemical composition of paired he-
2000. Aberrant splicing of tau pre-mRNA lical filaments in corticobasal degeneration.
caused by intronic mutations associated with Am. J. Pathol. 145(6):1496–508
the inherited dementia frontotemporal de- Kusakawa G, Saito T, Onuki R, Ishiguro K,
mentia with parkinsonism linked to chromo- Kishimoto T, Hisanaga S. 2000. Calpain-
some 17. Mol. Cell Biol. 20(11):4036–48 dependent proteolytic cleavage of the p35
Kampers T, Friedhoff P, Biernat J, Mandelkow cyclin-dependent kinase 5 activator to p25.
EM, Mandelkow E. 1996. RNA stimulates J. Biol. Chem. 275(22):17166–72
P1: FUM
April 7, 2001 15:58 Annual Reviews AR121-37

NEW INSIGHTS INTO TAUOPATHIES 1153

Lee G, Neve RL, Kosik KS. 1989. The micro- tion by cAMP-dependent protein kinase in-
tubule binding domain of tau protein. Neuron hibits the degradation of tau by calpain. J.
2(6):1615–24 Biol. Chem. 267(3):1563–68
Lee MS, Kwon YT, Li M, Peng J, Friedlander Litvan I, Agid Y, Calne D, Campbell G, Dubois
RM, Tsai LH. 2000. Neurotoxicity induces B, et al. 1996. Clinical research criteria
cleavage of p35 to p25 by calpain. Nature for the diagnosis of progressive supranu-
405(6784):360–64 clear palsy (Steele-Richardson-Olszewski
Lee VM-Y, Balin BJ, Otvos L Jr, Trojanowski syndrome): report of the NINDS-SPSP in-
JQ. 1991. A68: a major subunit of paired he- ternational workshop. Neurology 47(1):1–9
lical filaments and derivatized forms of nor- LoPresti P, Szuchet S, Papasozomenos SC,
mal tau. Science 251(4994):675–78 Zinkowski RP, Binder LI. 1995. Functional
Annu. Rev. Neurosci. 2001.24:1121-1159. Downloaded from www.annualreviews.org

Lendon CL, Lynch T, Norton J, McKeel DW Jr, implications for the microtubule-associated
Busfield F, et al. 1998. Hereditary dyspha- protein tau: localization in oligodendrocytes.
sic disinhibition dementia: a frontotempo- Proc. Natl. Acad. Sci. USA 92(22):10369–73
ral dementia linked to 17q21–22. Neurology Lovestone S, Davis DR, Webster MT, Kaech
50(6):1546–55 S, Brion JP, et al. 1999. Lithium reduces tau
by Open University on 07/11/13. For personal use only.

Leost M, Schultz C, Link A, Wu YZ, Biernat phosphorylation: effects in living cells and in
J, et al. 2000. Paullones are potent inhibitors neurons at therapeutic concentrations. Biol.
of glycogen synthase kinase-3β and cyclin- Psychiatry 45(8):995–1003
dependent kinase 5/p25. Eur. J. Biochem. Lovestone S, Hartley CL, Pearce J, Anderton
267(19):5983–94 BH. 1996. Phosphorylation of tau by glyco-
Lew J, Huang QQ, Qi Z, Winkfein RJ, Ae- gen synthase kinase-3 beta in intact mam-
bersold R, et al. 1994. A brain-specific ac- malian cells: the effects on the organization
tivator of cyclin-dependent kinase 5. Nature and stability of microtubules. Neuroscience
371(6496):423–26 73(4):1145–57
Lew J, Wang JH. 1994. Neuronal cdc2-like ki- Lu PJ, Wulf G, Zhou XZ, Davies P, Lu KP. 1999.
nase. Trends Biochem. Sci. 20:33–37 The prolyl isomerase Pin1 restores the func-
Lewis J, McGowan E, Rockwood J, Melrose H, tion of Alzheimer-associated phosphorylated
Nacharaju P, et al. 2000. Neurofibrillary tan- tau protein. Nature 399(6738):784–88
gles, amyotrophy and progressive motor dis- Lund and Manchester Groups. 1994. Clinical
turbance in mice expressing mutant (P301L) and neuropathological criteria for frontotem-
tau protein. Nat. Genet. 25(4):402–5 poral dementia. The Lund and Manchester
Liao H, Li Y, Brautigan DL, Gundersen GG. Groups. J. Neurol. Neurosurg. Psychiatry
1998. Protein phosphatase 1 is targeted to 57(4):416–18
microtubules by the microtubule-associated Mailliot C, Sergeant N, Bussière T, Caillet-
protein tau. J. Biol. Chem. 273(34):21901–8 Boudin ML, Delacourte A, Buée L. 1998.
Lieberman AP, Trojanowski JQ, Lee VM-Y, Phosphorylation of specific sets of tau iso-
Balin BJ, Ding XS, et al. 1998. Cognitive, forms reflects different neurofibrillary degen-
neuroimaging, and pathological studies in eration processes. FEBS Lett. 433(3):201–4
a patient with Pick’s disease. Ann. Neurol. Mandelkow EM, Drewes G, Biernat J, Gustke
43(2):259–65 N, Van Lint J, et al. 1992. Glycogen syn-
Lippa CF, Zhukareva V, Kawarai T, Uryu thase kinase-3 and the Alzheimer-like state
H, Shafiq M, et al. 2000. Frontotemporal of microtubule-associated protein tau. FEBS
dementia with novel tau pathology and a Lett. 314(3):315–21
Glu342Val tau gene mutation. Ann. Neurol. Mann DM. 1998. Dementia of frontal type
48(6):850–58 and dementias with subcortical gliosis. Brain
Litersky JM, Johnson GV. 1992. Phosphoryla- Pathol. 8(2):325–38
P1: FUM
April 7, 2001 15:58 Annual Reviews AR121-37

1154 LEE ¥ GOEDERT ¥ TROJANOWSKI

Matsumoto S, Hirano A, Goto S. 1990. diseases. J. Neurol. Neurosurg. Psychiatry


Spinal cord neurofibrillary tangles of 66(5):665–67
Guamanian amyotrophic lateral sclerosis Morris HR, Perez-Tur J, Janssen JC, Brown
and parkinsonism-dementia complex: an J, Lees AJ, et al. 1999b. Mutation in the
immunohistochemical study. Neurology tau exon 10 splice site region in famil-
40(6):975–79 ial frontotemporal dementia. Ann. Neurol.
Matsumura N, Yamazaki T, Ihara Y. 1999. 45(2):270–71
Stable expression in Chinese hamster ovary Munoz-Garcia D, Ludwin SK. 1984. Classic
cells of mutated tau genes causing frontotem- and generalized variants of Pick’s disease: a
poral dementia and parkinsonism linked to clinicopathological, ultrastructural, and im-
chromosome 17 (FTDP-17). Am. J. Pathol. munocytochemical comparative study. Ann.
Annu. Rev. Neurosci. 2001.24:1121-1159. Downloaded from www.annualreviews.org

154(6):1649–56 Neurol. 16(4):467–80


Matsuo ES, Shin RW, Billingsley ML, Van de- Munoz-Montano JR, Moreno FJ, Avila J, Diaz-
Voorde A, O’Connor M, et al. 1994. Biopsy- Nido J. 1997. Lithium inhibits Alzheimer’s
derived adult human brain tau is phosphory- disease-like tau protein phosphorylation
lated at many of the same sites as Alzheimer’s in neurons. FEBS Lett. 411(2–3):183–
by Open University on 07/11/13. For personal use only.

disease paired helical filament tau. Neuron 88


13(4):989–1002 Murayama S, Mori H, Ihara Y, Tomonaga M.
Mawal-Dewan M, Henley J, Van de Voorde A, 1990. Immunocytochemical and ultrastruc-
Trojanowski JQ, Lee VM-Y. 1994. The phos- tural studies of Pick’s disease. Ann. Neurol.
phorylation state of tau in the developing rat 27(4):394–405
brain is regulated by phosphoprotein phos- Murrell JR, Koller D, Foroud T, Goedert
phatases. J. Biol. Chem. 269(49):30981–87 M, Spillantini MG, et al. 1997. Familial
Merrick SE, Trojanowski JQ, Lee VM-Y. 1997. multiple-system tauopathy with presenile de-
Selective destruction of stable microtubules mentia is localized to chromosome 17. Am.
and axons by inhibitors of protein ser- J. Hum. Genet. 61(5):1131–38
ine/threonine phosphatases in cultured hu- Murrell JR, Spillantini MG, Zolo P, Guazzelli
man neurons. J. Neurosci. 17(15):5726–37 M, Smith MJ, et al. 1999. Tau gene muta-
Mirra SS, Murrell JR, Gearing M, Spillantini tion G389R causes a tauopathy with abun-
MG, Goedert M, et al. 1999. Tau pathology dant Pick body-like inclusions and ax-
in a family with dementia and a P301L mu- onal deposits. J. Neuropathol. Exp. Neurol.
tation in tau. J. Neuropathol. Exp. Neurol. 58(12):1207–26
58(4):335–45 Nacharaju P, Lewis J, Easson C, Yen S, Hackett
Mori H, Nishimura M, Namba Y, Oda M. 1994. J, et al. 1999. Accelerated filament formation
Corticobasal degeneration: a disease with from tau protein with specific FTDP-17 mis-
widespread appearance of abnormal tau and sense mutations. FEBS Lett. 447(2–3):195–
neurofibrillary tangles, and its relation to 99
progressive supranuclear palsy. Acta Neu- Nasreddine ZS, Loginov M, Clark LN,
ropathol. 88(2):113–21 Lamarche J, Miller BL, et al. 1999. From
Morishima-Kawashima M, Hasegawa M, Takio genotype to phenotype: a clinical patho-
K, Suzuki M, Yoshida H, et al. 1995. Hy- logical, and biochemical investigation of
perphosphorylation of tau in PHF. Neurobiol. frontotemporal dementia and parkinsonism
Aging 16(3):365–71 (FTDP-17) caused by the P301L tau muta-
Morris HR, Janssen JC, Bandmann O, Daniel tion. Ann. Neurol. 45(6):704–15
SE, Rossor MN, et al. 1999a. The tau gene Nath R, Davis M, Probert AW, Kupina NC, Ren
A0 polymorphism in progressive supranu- X, et al. 2000. Processing of cdk5 activator
clear palsy and related neurodegenerative p35 to its truncated form (p25) by calpain
P1: FUM
April 7, 2001 15:58 Annual Reviews AR121-37

NEW INSIGHTS INTO TAUOPATHIES 1155

in acutely injured neuronal cells. Biochem. Prusiner SB. 1998. Prions. Proc. Natl. Acad.
Biophys. Res. Commun. 274(1):16–21 Sci. USA 95(23):13363–83
Neve RL, Harris P, Kosik KS, Kurnit DM, Rebeiz J, Kolodny EH, Richardson EP Jr. 1967.
Donlon TA. 1986. Identification of cDNA Corticodentatonigral degeneration with neu-
clones for the human microtubule-associated ronal achromasia: a progressive disorder in
protein tau and chromosomal localization of late adult life. Trans. Am. Neurol. Assoc.
the genes for tau and microtubule-associated 92:23–26
protein 2. Brain Res. 387(3):271–80 Rebeiz JJ, Kolodny EH, Richardson EP
Patrick GN, Zukerberg L, Nikolic M, de la Jr. 1968. Corticodentatonigral degeneration
Monte S, Dikkes P, Tsai LH. 1999. Conver- with neuronal achromasia 2. Arch. Neurol.
sion of p35 to p25 deregulates Cdk5 activ- 18(1):20–33
Annu. Rev. Neurosci. 2001.24:1121-1159. Downloaded from www.annualreviews.org

ity and promotes neurodegeneration. Nature Reed LA, Schmidt ML, Wszolek ZK, Balin BJ,
402(6762):615–22 Soontornniyomkij V, et al. 1998. The neu-
Perez M, Lim F, Arrasate M, Avila J. 2000. The ropathology of a chromosome 17-linked au-
FTDP-17-linked mutation R406W abolishes tosomal dominant parkinsonism and demen-
the interaction of phosphorylated tau with tia (“pallido-ponto-nigral degeneration”). J.
by Open University on 07/11/13. For personal use only.

microtubules. J. Neurochem. 74(6):2583–89 Neuropathol. Exp. Neurol. 57(6):588–601


Perez M, Valpuesta JM, Medina M, Montejo G, Rep. Work Group Frontotemporal Dementia
Avila J. 1996. Polymerization of tau into fil- Pick’s Disease. 2001. Clinical and neu-
aments in the presence of heparin: the mini- ropathological diagnosis of frontotemporal
mal sequence required for tau-tau interaction. dementias. Neurobiol. Aging. In press
J. Neurochem. 67(3):1183–90 Reynolds CH, Nebreda AR, Gibb GM, Utton
Pickering-Brown S, Baker M, Yen SH, Liu WK, MA, Anderton BH. 1997a. Reactivating ki-
Hasegawa M, et al. 2000. Pick’s disease is as- nase/p38 phosphorylates tau protein in vitro.
sociated with mutations in the tau gene. Ann. J. Neurochem. 69(1):191–98
Neurol. 48(5):806–8 Reynolds CH, Utton MA, Gibb GM, Yates A,
Pollock NJ, Mirra SS, Binder LI, Hansen Anderton BH. 1997b. Stress-activated pro-
LA, Wood JG. 1986. Filamentous aggre- tein kinase/c-jun N-terminal kinase phos-
gates in Pick’s disease, progressive supranu- phorylates tau protein. J. Neurochem.
clear palsy, and Alzheimer’s disease share 68(4):1736–44
antigenic determinants with microtubule- Rizzini C, Goedert M, Hodges JR, Smith
associated protein, tau. Lancet 2(8517): MJ, Jakes R, et al. 2000. Tau gene muta-
1211 tion K257T causes a tauopathy similar to
Poorkaj P, Bird TD, Wijsman E, Nemens E, Gar- Pick’s disease. J. Neuropathol. Exp. Neurol.
ruto RM, et al. 1998. Tau is a candidate gene 59(11):990–1001
for chromosome 17 frontotemporal demen- Rizzu P, Joose M, Ravid R, Hoogeveen A, Kam-
tia. Ann. Neurol. 43(6):815–25 phorst W, et al. 2000. Mutation-dependent
Probst A, Götz J, Wiederhold KH, Tolnay aggregation of tau protein and its selective
M, Mistl C, et al. 2000. Axonopathy and depletion from the soluble fraction in brain of
amyotrophy in mice transgenic for human P301L FTDP-17 patients. Hum. Mol. Genet.
four-repeat tau protein. Acta Neuropathol. 9(20):3075–82
99(5):469–81 Rizzu P, Van Swieten JC, Joosse M, Hasegawa
Probst A, Tolnay M, Langui D, Goedert M, M, Stevens M, et al. 1999. High prevalence of
Spillantini MG. 1996. Pick’s disease: hy- mutations in the microtubule-associated pro-
perphosphorylated tau protein segregates to tein tau in a population study of frontotempo-
the somatoaxonal compartment. Acta Neu- ral dementia in the Netherlands. Am. J. Hum.
ropathol. 92(6):588–96 Genet. 64(2):414–21
P1: FUM
April 7, 2001 15:58 Annual Reviews AR121-37

1156 LEE ¥ GOEDERT ¥ TROJANOWSKI

Roy S, Datta CK, Hirano A, Ghatak NR, Zim- paired helical filament tau. J. Biol. Chem.
merman HM. 1974. Electron microscopic 270(32):18917–22
study of neurofibrillary tangles in Steele- Shin RW, Iwaki T, Kitamoto T, Tateishi J. 1991.
Richardson-Olszewski syndrome. Acta Neu- Hydrated autoclave pretreatment enhances
ropathol. 29(2):175–79 tau immunoreactivity in formalin-fixed nor-
Sahara N, Tomiyama T, Mori H. 2000. Mis- mal and Alzheimer’s disease brain tissues.
sense point mutations of tau to segregate with Lab. Invest. 64(5):693–702
FTDP-17 exhibit site-specific effects on mi- Si ZH, Rauch D, Stoltzfus CM. 1998. The
crotubule structure in COS cells: a novel ac- exon splicing silencer in human immunode-
tion of R406W mutation. J. Neurosci. Res. ficiency virus type 1 Tat exon 3 is bipartite
60(3):380–87 and acts early in spliceosome assembly. Mol.
Annu. Rev. Neurosci. 2001.24:1121-1159. Downloaded from www.annualreviews.org

Schmidt ML, Huang R, Martin JA, Henley J, Cell Biol. 18(9):5404–13


Mawal-Dewan M, et al. 1996. Neurofibril- Singh TJ, Zaidi T, Grundke-Iqbal I, Iqbal K.
lary tangles in progressive supranuclear palsy 1995. Modulation of GSK-3-catalyzed phos-
contain the same tau epitopes identified in phorylation of microtubule-associated pro-
Alzheimer’s disease PHFtau. J. Neuropathol. tein tau by nonproline-dependent protein ki-
by Open University on 07/11/13. For personal use only.

Exp. Neurol. 55(5):534–39 nases. FEBS Lett. 358(1):4–8


Schneider A, Biernat J, von Bergen M, Snow AD, Mar H, Nochlin D, Sekiguchi RT,
Mandelkow E, Mandelkow EM. 1999. Kimata K, et al. 1990. Early accumulation
Phosphorylation that detaches tau pro- of heparan sulfate in neurons and in the
tein from microtubules (Ser262, Ser214) beta-amyloid protein-containing lesions of
also protects it against aggregation into Alzheimer’s disease and Down’s syndrome.
Alzheimer paired helical filaments. Biochem- Am. J. Pathol. 137(5):1253–70
istry 38(12):3549–58 Sobue K, Agarwal-Mawal A, Wei L, Sun W,
Schweers O, Schönbrunn-Hanebeck E, Marx Miura Y, Paudel HK. 2000. Interaction of
A, Mandelkow E. 1994. Structural studies of neuronal cdc2 like protein kinase with micro-
tau protein and Alzheimer paired helical fil- tubule associated protein tau. J. Biol. Chem.
aments show no evidence for beta-structure. 275(22):16673–80
J. Biol. Chem. 269(39):24290–97 Sontag E, Nunbhakdi-Craig V, Bloom GS,
Sengupta A, Wu Q, Grundke-Iqbal I, Iqbal Mumby MC. 1995. A novel pool of pro-
K, Singh TJ. 1997. Potentiation of GSK- tein phosphatase 2A is associated with mi-
3-catalyzed Alzheimer-like phosphorylation crotubules and is regulated during the cell
of human tau by cdk5. Mol. Cell Biochem. cycle. J. Cell Biol. 128(6):1131–44
167(1–2):99–105 Sontag E, Nunbhakdi-Craig V, Lee G, Brandt
Sergeant N, David JP, Lefranc D, Vermersch P, R, Kamibayashi C, et al. 1999. Molecular
Wattez A, Delacourte A. 1997. Different dis- interactions among protein phosphatase 2A,
tribution of phosphorylated tau protein iso- tau, and microtubules. Implications for the
forms in Alzheimer’s and Pick’s diseases. regulation of tau phosphorylation and the
FEBS Lett. 412(3):578–82 development of tauopathies. J. Biol. Chem.
Sergeant N, Wattez A, Delacourte A. 1999. 274(36):25490–98
Neurofibrillary degeneration in progressive Sperfeld AD, Collatz MB, Baier H, Palm-
supranuclear palsy and corticobasal degener- bach M, Storch A, et al. 1999. FTDP-17:
ation: tau pathologies with exclusively “exon an early-onset phenotype with parkinsonism
10” isoforms. J. Neurochem. 72(3):1243–49 and epileptic seizures caused by a novel mu-
Seubert P, Mawal-Dewan M, Barbour R, Jakes tation. Ann. Neurol. 46(5):708–15
R, Goedert M, et al. 1995. Detection of phos- Spillantini MG, Bird TD, Ghetti B. 1998a.
phorylated Ser262 in fetal tau, adult tau, and Frontotemporal dementia and parkinsonism
P1: FUM
April 7, 2001 15:58 Annual Reviews AR121-37

NEW INSIGHTS INTO TAUOPATHIES 1157

linked to chromosome 17: a new group of type caused by tau gene mutations. Brain
tauopathies. Brain Pathol. 8(2):387–402 123(5):880–93
Spillantini MG, Crowther RA, Goedert M. Steele JC, Richardson JC, Olszewski J. 1964.
1996. Comparison of the neurofibrillary Progressive supranuclear palsy. Arch. Neu-
pathology in Alzheimer’s disease and fa- rol. 10:333–59
milial presenile dementia with tangles. Acta Sturchler-Pierrat C, Abramowski D, Duke
Neuropathol. 92(1):42–48 M, Wiederhold KH, Mistl C, et al. 1997.
Spillantini MG, Crowther RA, Kamphorst W, Two amyloid precursor protein transgenic
Heutink P, Van Swieten JC. 1998b. Tau mouse models with Alzheimer disease-
pathology in two Dutch families with mu- like pathology. Proc. Natl. Acad. Sci. USA
tations in the microtubule-binding region of 94(24):13287–92
Annu. Rev. Neurosci. 2001.24:1121-1159. Downloaded from www.annualreviews.org

tau. Am. J. Pathol. 153(5):1359–63 Takahashi M, Tomizawa K, Ishiguro K, Taka-


Spillantini MG, Goedert M, Crowther RA, Mur- matsu M, Fujita SC, Imahori K. 1995. In-
rell JR, Farlow MR, Ghetti B. 1997. Famil- volvement of tau protein kinase I in paired
ial multiple system tauopathy with presenile helical filament-like phosphorylation of the
dementia: a disease with abundant neuronal juvenile tau in rat brain. J. Neurochem.
by Open University on 07/11/13. For personal use only.

and glial tau filaments. Proc. Natl. Acad. Sci. 64(4):1759–68


USA 94(8):4113–18 Takahashi M, Yasutake K, Tomizawa K. 1999.
Spillantini MG, Murrell JR, Goedert M, Far- Lithium inhibits neurite growth and tau pro-
low MR, Klug A, Ghetti B. 1998c. Mu- tein kinase I/glycogen synthase kinase-3β-
tation in the tau gene in familial multiple dependent phosphorylation of juvenile tau
system tauopathy with presenile dementia. in cultured hippocampal neurons. J. Neu-
Proc. Natl. Acad. Sci. USA 95(13):7737– rochem. 73(5):2073–83
41 Tanaka K, Watakabe A, Shimura Y. 1994. Poly-
Spillantini MG, Yoshida H, Rizzini C, Lantos purine sequences within a downstream exon
PL, Khan N, et al. 2000. A novel tau mutation function as a splicing enhancer. Mol. Cell
(N296N) in familial dementia with swollen Biol. 14(2):1347–54
achromatic neurons and corticobasal inclu- Tellez-Nagel I, Wisniewski HM. 1973. Ul-
sion bodies. Ann. Neurol. 48(6):939–43 trastructure of neurofibrillary tangles in
Spittaels K, Van den Haute C, Van Dorpe J, Steele-Richardson-Olszewski syndrome.
Bruynseels K, Vandezande K, et al. 1999. Arch. Neurol. 29(5):324–27
Prominent axonopathy in the brain and spinal Tesseur I, Van Dorpe J, Spittaels K, Van den
cord of transgenic mice overexpressing four- Haute C, Moechars D, Van Leuven F. 2000.
repeat human tau protein. Am. J. Pathol. Expression of human apolipoprotein E4 in
155(6):2153–65 neurons causes hyperphosphorylation of pro-
Spittaels K, Van den Haute C, Van Dorpe J, tein tau in the brains of transgenic mice. Am.
Geerts H, Mercken M, et al. 2000. Glyco- J. Pathol. 156(3):951–64
gen synthase kinase-3beta phosphorylates Tolnay M, Spillantini MG, Rizzini C, Eccles
protein tau and rescues the axonopathy in D, Lowe J, Ellison D. 2000. A new case of
the central nervous system of human four- frontotemporal dementia and parkinsonism
repeat tau transgenic mice. J. Biol. Chem. resulting from an intron 10 +3-splice site
275(52):41340–49 mutation in the tau gene: clinical and patho-
Stanford PM, Halliday GM, Brooks WS, Kwok logical features. Neuropathol. Appl. Neuro-
JB, Storey CE, et al. 2000. Progressive biol. 26(4):368–78
supranuclear palsy pathology caused by a Trojanowski JQ, Goedert M, Iwatsubo T,
novel silent mutation in exon 10 of the Lee VM-Y. 1998. Fatal attraction: abnor-
tau gene: expansion of the disease pheno- mal protein aggregation and neuron death in
P1: FUM
April 7, 2001 15:58 Annual Reviews AR121-37

1158 LEE ¥ GOEDERT ¥ TROJANOWSKI

Parkinson’s disease and Lewy body demen- Lansberg C, Zhang B, Hong M, et al. 2000.
tia. Cell Death Differ. 5:832–37 Distinct FTDP-17 missense mutations in tau
Trojanowski JQ, Lee VM-Y. 1994. Paired produce tau aggregates and other phatholo-
helical filament tau in Alzheimer’s dis- gical phenotypes in transfected CHO cells.
ease. The kinase connection. Am. J. Pathol. Mol. Biol. Cell. 11(12):4093–104
144(3):449–53 von Bergen M, Friedhoff P, Biernat J, Heberle
Tsai LH, Delalle I, Caviness VS Jr, Chae T, J, Mandelkow EM, Mandelkow E. 2000. As-
Harlow E. 1994. p35 is a neural-specific reg- sembly of tau protein into Alzheimer paired
ulatory subunit of cyclin-dependent kinase 5. helical filaments depends on a local se-
Nature 371(6496):419–23 quence motif ((306)VQIVYK(311)) forming
Tseng HC, Graves DJ. 1998. Natural methyl- beta structure. Proc. Natl. Acad. Sci. USA
Annu. Rev. Neurosci. 2001.24:1121-1159. Downloaded from www.annualreviews.org

amine osmolytes, trimethylamine N-oxide 97(10):5129–34


and betaine, increase tau-induced polymer- Watanabe A, Hasegawa M, Suzuki M, Takio
ization of microtubules. Biochem. Biophys. K, Morishima-Kawashima M, et al. 1993. In
Res. Commun. 250(3):726–30 vivo phosphorylation sites in fetal and adult
Tseng HC, Lu Q, Henderson E, Graves DJ. rat tau. J. Biol. Chem. 268(34):25712–17
by Open University on 07/11/13. For personal use only.

1999. Phosphorylated tau can promote tubu- Weingarten MD, Lockwood AH, Hwo SY,
lin assembly. Proc. Natl. Acad. Sci. USA Kirschner MW. 1975. A protein factor es-
96(17):9503–8 sential for microtubule assembly. Proc. Natl.
Van Swieten JC, Stevens M, Rosso SM, Rizzu P, Acad. Sci. USA 72(5):1858–62
Joosse M, et al. 1999. Phenotypic variation Wijker M, Wszolek ZK, Wolters EC, Rooimans
in hereditary frontotemporal dementia with MA, Pals G, et al. 1996. Localization of
tau mutations. Ann. Neurol. 46(4):617–26 the gene for rapidly progressive autosomal
Varani L, Hasegawa M, Spillantini MG, Smith dominant parkinsonism and dementia with
MJ, Murrell JR, et al. 1999. Structure of pallido-ponto-nigral degeneration to chro-
tau exon 10 splicing regulatory element mosome 17q21. Hum. Mol. Genet. 5(1):151–
RNA and destabilization by mutations of 54
frontotemporal dementia and parkinsonism Wilhelmsen KC, Lynch T, Pavlou E, Hig-
linked to chromosome 17. Proc. Natl. Acad. gins M, Nygaard TG. 1994. Localization
Sci. USA 96(14):8229–34 of disinhibition-dementia-parkinsonism-
Varani L, Spillantini MG, Goedert M, Varani G. amyotrophy complex to 17q21–22. Am. J.
2000. Structural basis for recognition of the Hum. Genet. 55(6):1159–65
RNA major groove in the tau exon 10 splic- Wille H, Drewes G, Biernat J, Mandelkow EM,
ing regulatory element by aminoglycoside Mandelkow E. 1992. Alzheimer-like paired
antibiotics. Nucleic Acids Res. 28(3):710–19 helical filaments and antiparallel dimers
Verbeek MM, Otte-Holler I, van den Born J, van formed from microtubule-associated protein
den Heuvel LP, David G, et al. 1999. Agrin tau in vitro. J. Cell Biol. 118(3):573–84
is a major heparan sulfate proteoglycan accu- Wilson DM, Binder LI. 1997. Free fatty
mulating in Alzheimer’s disease brain. Am. J. acids stimulate the polymerization of tau
Pathol. 155(6):2115–25 and amyloid beta peptides. In vitro evi-
Vermersch P, Robitaille Y, Bernier L, Wattez dence for a common effector of pathogen-
A, Gauvreau D, Delacourte A. 1994. Bio- esis in Alzheimer’s disease. Am. J. Pathol.
chemical mapping of neurofibrillary degen- 150(6):2181–95
eration in a case of progressive supranuclear Wischik CM, Novak M, Thogersen HC, Ed-
palsy: evidence for general cortical involve- wards PC, Runswick MJ, et al. 1988. Iso-
ment. Acta Neuropathol. 87(6):572–77 lation of a fragment of tau derived from
Vogelsberg-Ragaglia V, Bruce J, Richter- the core of the paired helical filament of
P1: FUM
April 7, 2001 15:58 Annual Reviews AR121-37

NEW INSIGHTS INTO TAUOPATHIES 1159

Alzheimer disease. Proc. Natl. Acad. Sci. tation at posititon +12 in the intron fol-
USA 85(12):4506–10 lowing exon 10 of the tau gene in familial
Woody RW, Clark DC, Roberts GC, Martin SR, frontotemporal dementia (FTD-Kumamoto).
Bayley PM. 1983. Molecular flexibility in Ann. Neurol. 47(4):422–29
microtubule proteins: proton nuclear mag- Yen SH, Hutton M, DeTure M, Ko LW,
netic resonance characterization. Biochem- Nacharaju P. 1999. Fibrillogenesis of tau: in-
istry 22(9):2186–92 sights from tau missense mutations in FTDP-
Yamada T, McGeer PL, McGeer EG. 1992. 17. Brain Pathol. 9(4):695–705
Appearance of paired nucleated, tau-positive Yoshida H, Ihara Y. 1993. Tau in paired helical
glia in patients with progressive supranuclear filaments is functionally distinct from fetal
palsy brain tissue. Neurosci. Lett. 135(1):99– tau: assembly incompetence of paired helical
Annu. Rev. Neurosci. 2001.24:1121-1159. Downloaded from www.annualreviews.org

102 filament-tau. J. Neurochem. 61(3):1183–86


Yamaguchi H, Ishiguro K, Uchida T, Takashima Zheng-Fischhöfer Q, Biernat J, Mandelkow
A, Lemere CA, Imahori K. 1996. Preferential EM, Illenberger S, Godemann R, Mandelkow
labeling of Alzheimer neurofibrillary tangles E. 1998. Sequential phosphorylation of tau
with antisera for tau protein kinase (TPK) by glycogen synthase kinase-3β and protein
by Open University on 07/11/13. For personal use only.

I/glycogen synthase kinase-3 beta and cyclin- kinase A at Thr212 and Ser214 gener-
dependent kinase 5, a component of TPK II. ates the Alzheimer-specific epitope of anti-
Acta Neuropathol. 92(3):232–41 body AT100 and requires a paired-helical-
Yasuda M, Kawamata T, Komure O, Kuno filament-like conformation. Eur. J. Biochem.
S, D’Souza I, et al. 1999. A mutation in 252(3):542–52
the microtubule-associated protein tau in Zhukareva V, Vogelsberg-Ragaglia V, Van
pallido-nigro-luysian degeneration. Neurol- Deerlin VMD, Bruce J, Schuck T, et al. 2001.
ogy 53(4):864–68 Loss of brain tau defines novel sporadic and
Yasuda M, Takamatsu J, D’Souza I, Crowther familial tauopathies with frontotemporal de-
RA, Kawamata T, et al. 2000. A novel mu- mentia. Ann. Neurol. 49(1):165–75
Annual Review of Neuroscience
Volume 24, 2001

CONTENTS
PDZ DOMAINS AND THE ORGANIZATION OF
1
SUPRAMOLECULAR COMPLEXES, Morgan Sheng, Carlo Sala

THE ROLE AND REGULATION OF ADENOSINE IN THE CENTRAL 31


NERVOUS SYSTEM, Thomas V. Dunwiddie, Susan A. Masino
LOCALIZATION AND GLOBALIZATION IN CONSCIOUS VISION,
57
S. Zeki
GLIAL CONTROL OF NEURONAL DEVELOPMENT, Greg Lemke 87
TOUCH AND GO: Decision-Making Mechanisms in Somatosensation,
107
Ranulfo Romo, Emilio Salinas
SYNAPTIC MODIFICATION BY CORRELATED ACTIVITY: Hebb''s
139
Annu. Rev. Neurosci. 2001.24:1121-1159. Downloaded from www.annualreviews.org

Postulate Revisited, Guo-qiang Bi, Mu-ming Poo


AN INTEGRATIVE THEORY OF PREFRONTAL CORTEX
167
FUNCTION, Earl K. Miller, Jonathan D. Cohen
THE PHYSIOLOGY OF STEREOPSIS, B. G. Cumming, G. C.
203
by Open University on 07/11/13. For personal use only.

DeAngelis
PARANEOPLASTIC NEUROLOGIC DISEASE ANTIGENS: RNA-
Binding Proteins and Signaling Proteins in Neuronal Degeneration, Kiran 239
Musunuru, Robert B. Darnell
ODOR ENCODING AS AN ACTIVE, DYNAMICAL PROCESS:
Experiments, Computation, and Theory, Gilles Laurent, Mark Stopfer,
263
Rainer W Friedrich, Misha I Rabinovich, Alexander Volkovskii, Henry
DI Abarbanel
PROTEIN SYNTHESIS AT SYNAPTIC SITES ON DENDRITES,
299
Oswald Steward, Erin M. Schuman

SIGNALING AND TRANSCRIPTIONAL MECHANISMS IN 327


PITUITARY DEVELOPMENT, Jeremy S. Dasen, Michael G. Rosenfeld
NEUROPEPTIDES AND THE INTEGRATION OF MOTOR
357
RESPONSES TO DEHYDRATION , Alan G. Watts
THE DEVELOPMENTAL BIOLOGY OF BRAIN TUMORS, Robert
385
Wechsler-Reya, Matthew P. Scott
TO EAT OR TO SLEEP? OREXIN IN THE REGULATION OF
FEEDING AND WAKEFULNESS, Jon T. Willie, Richard M. Chemelli, 429
Christopher M. Sinton, Masashi Yanagisawa
SPATIAL PROCESSING IN THE BRAIN: The Activity of Hippocampal
459
Place Cells, Phillip J. Best, Aaron M. White, Ali Minai
THE VANILLOID RECEPTOR: A Molecular Gateway to the Pain
487
Pathway, Michael J Caterina, David Julius
PRION DISEASES OF HUMANS AND ANIMALS: Their Causes and
519
Molecular Basis, John Collinge
VIKTOR HAMBURGER AND RITA LEVI-MONTALCINI: The Path to
551
the Discovery of Nerve Growth Factor, W. Maxwell Cowan
EARLY DAYS OF THE NERVE GROWTH FACTOR PROTEINS,
601
Eric M. Shooter
SEQUENTIAL ORGANIZATION OF MULTIPLE MOVEMENTS:
631
Involvement of Cortical Motor Areas, Jun Tanji
INFLUENCE OF DENDRITIC CONDUCTANCES ON THE INPUT-
653
OUTPUT PROPERTIES OF NEURONS, Alex Reyes
NEUROTROPHINS: Roles in Neuronal Development and Function, Eric
677
J Huang, Louis F Reichardt
CONTRIBUTIONS OF THE MEDULLARY RAPHE AND
VENTROMEDIAL RETICULAR REGION TO PAIN MODULATION 737
AND OTHER HOMEOSTATIC FUNCTIONS, Peggy Mason
ACTIVATION, DEACTIVATION, AND ADAPTATION IN
VERTEBRATE PHOTORECEPTOR CELLS, Marie E Burns, Denis A
Baylor 779
ACTIVITY-DEPENDENT SPINAL CORD PLASTICITY IN HEALTH
AND DISEASE, Jonathan R Wolpaw, Ann M Tennissen 807
QUANTITATIVE GENETICS AND MOUSE BEHAVIOR, Jeanne M
845
Wehner, Richard A Radcliffe, Barbara J Bowers
EARLY ANTERIOR/POSTERIOR PATTERNING OF THE
869
MIDBRAIN AND CEREBELLUM, Aimin Liu, Alexandra L Joyner
NEUROBIOLOGY OF PAVLOVIAN FEAR CONDITIONING, Stephen
897
Maren
{{alpha}}-LATROTOXIN AND ITS RECEPTORS: Neurexins and
Annu. Rev. Neurosci. 2001.24:1121-1159. Downloaded from www.annualreviews.org

CIRL/Latrophilins, Thomas C Südhof 933


IMAGING AND CODING IN THE OLFACTORY SYSTEM, John S
963
Kauer, Joel White
THE ROLE OF THE CEREBELLUM IN VOLUNTARY EYE
by Open University on 07/11/13. For personal use only.

981
MOVEMENTS, Farrel R Robinson, Albert F Fuchs
ROLE OF THE REELIN SIGNALING PATHWAY IN CENTRAL
1005
NERVOUS SYSTEM DEVELOPMENT, Dennis S Rice, Tom Curran
HUMAN BRAIN MALFORMATIONS AND THEIR LESSONS FOR
1041
NEURONAL MIGRATION, M Elizabeth Ross, Christopher A Walsh
MORPHOLOGICAL CHANGES IN DENDRITIC SPINES
ASSOCIATED WITH LONG-TERM SYNAPTIC PLASTICITY, Rafael 1071
Yuste, Tobias Bonhoeffer

STOPPING TIME: The Genetics of Fly and Mouse Circadian Clocks, 1091
Ravi Allada, Patrick Emery, Joseph S. Takahashi, Michael Rosbash
NEURODEGENERATIVE TAUOPATHIES, Virginia M-Y Lee, Michel
1121
Goedert, John Q Trojanowski
MATERNAL CARE, GENE EXPRESSION, AND THE
TRANSMISSION OF INDIVIDUAL DIFFERENCES IN STRESS 1161
REACTIVITY ACROSS GENERATIONS, Michael J Meaney
NATURAL IMAGE STATISTICS AND NEURAL
1193
REPRESENTATION, Eero P Simoncelli, Bruno A Olshausen
Nerve Growth Factor Signaling, Neuroprotection, and Neural Repair,
1217
Michael V Sofroniew, Charles L Howe, William C Mobley
FLIES, GENES, AND LEARNING, Scott Waddell, William G Quinn 1283

You might also like