Virginia M-Y Lee,1 Michel Goedert,2 and John Q Trojanowski

41
Annu. Rev. Neurosci. 2001. 24:1121–159 Copyright c 2001 by Annual Reviews. All rights reserved NEURODEGENERATIVE T AUOPATHIES Virginia M-Y Lee, 1 Michel Goedert, 2 and John Q Trojanowski 1 1 Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104; e-mail: [email protected], [email protected] 2 Medical Research Council, Laboratory of Molecular Biology, Hills Road, Cambridge, CB2 2QH, United Kingdom; e-mail: [email protected] 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 Annu. Rev. Neurosci. 2001.24:1121-1159. Downloaded from www.annualreviews.org by Open University on 07/11/13. For personal use only.

Transcript of Virginia M-Y Lee,1 Michel Goedert,2 and John Q Trojanowski

Page 1: Virginia M-Y Lee,1 Michel Goedert,2 and John Q Trojanowski

P1: FUM

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

Annu. Rev. Neurosci. 2001. 24:1121–159Copyright c© 2001 by Annual Reviews. All rights reserved

NEURODEGENERATIVE TAUOPATHIES

Virginia M-Y Lee,1 Michel Goedert,2

and John Q Trojanowski11Center for Neurodegenerative Disease Research, Department of Pathology andLaboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia,Pennsylvania 19104; e-mail: [email protected],[email protected] Research Council, Laboratory of Molecular Biology, Hills Road, Cambridge,CB2 2QH, United Kingdom; e-mail: [email protected]

Key Words Alzheimer’s disease, frontotemporal dementia, mutation,neurodegenerative disease, filamentous deposits, pathology, tau protein

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

INTRODUCTION

The study of sporadic and familial neurodegenerative diseases over the past decadehas led to the realization that many of these disorders are characterized by distincthallmark 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

Ann

u. R

ev. N

euro

sci.

2001

.24:

1121

-115

9. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by O

pen

Uni

vers

ity o

n 07

/11/

13. F

or p

erso

nal u

se o

nly.

Page 2: Virginia M-Y Lee,1 Michel Goedert,2 and John Q Trojanowski

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-associatedprotein tau appears to share common mechanisms of disease. They are collectivelyknown as neurodegenerative tauopathies (Table 1). Despite their diverse pheno-typic manifestations, brain dysfunction and degeneration in tauopathies is linkedto the progressive accumulation of filamentous tau inclusions, and this, togetherwith the absence of other disease-specific neuropathological abnormalities, pro-vided circumstantial evidence implicating abnormal tau in disease onset and/orprogression. However, this view remained unproven and highly controversial un-til 1998, when multipletau gene mutations were discovered in frontotemporaldementia and parkinsonism linked to chromosome 17 (FTDP-17), thereby pro-viding unequivocal evidence that tau abnormalities alone are sufficient to causeneurodegenerative disease (Foster et al 1997, Hutton et al 1998, Poorkaj et al1998, Spillantini et al 1998c). This seminal finding opened up new avenues for

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-Str¨aussler-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

aDiseases in which tau-positive neurofibrillary pathology are the most predominant neu-ropathologic feature.

Ann

u. R

ev. N

euro

sci.

2001

.24:

1121

-115

9. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by O

pen

Uni

vers

ity o

n 07

/11/

13. F

or p

erso

nal u

se o

nly.

Page 3: Virginia M-Y Lee,1 Michel Goedert,2 and John Q Trojanowski

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 anddegeneration.

This review is designed to integrate and interpret the remarkable recent advancesthat have led to new insights into the mechanistic role that tau abnormalities playin neurodegenerative disease. It begins with a brief summary of our current under-standing of the humantau gene and the functions of the six alternatively splicedtau isoforms that are expressed in the normal adult human brain. What is knownabout 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 novelsporadic, tauopathies. This information provides the context and perspective inwhich to present an update on the increasing number of pathogenictau gene mu-tations, as well as a summary of the neuropathological and biochemical profiles ofthe tau pathologies in FTDP-17. Finally, the review concludes with an update ofthe current status of efforts to develop transgenic (TG) and other animal modelsof human neurodegenerative tauopathies.

STRUCTURE, FUNCTION, AND MOLECULARGENETICS OF TAU

Tau proteins are lowMr microtubule-associated proteins that are abundant in thecentral nervous system (CNS), where they are expressed predominantly in axons.They are also expressed in axons of peripheral nervous system neurons but arebarely 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). Humantau proteins are encoded by a single gene consisting of 16 exons on chromosome17q21, and the CNS isoforms are generated by alternative mRNA splicing of 11 ofthese 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 generatessix tau isoforms ranging from 352 to 441 amino acids in length (Figure 1), whichdiffer by the presence of either three (3R-tau) or four (4R-tau) carboxy-terminaltandem 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 splicingof 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 adulthuman brain, the ratio of 3R-tau to 4R-tau isoforms is∼1, but the 1N, 0N, and2N tau isoforms comprise about 54%, 37%, and 9%, respectively, of total tau(Goedert & Jakes 1990, Hong et al 1998). In addition, the alternative splicing oftau is developmentally regulated such that only the shortest tau isoform (3R/0N) isexpressed in fetal brain, whereas all six isoforms appear in the postnatal period ofthe human brain (Goedert et al 1989a). In the peripheral nervous system, inclusionof E4a in the amino-terminal half results in the expression of higherMr proteinstermed big tau (Georgieff et al 1991, Couchie et al 1992, Goedert et al 1992c).

Ann

u. R

ev. N

euro

sci.

2001

.24:

1121

-115

9. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by O

pen

Uni

vers

ity o

n 07

/11/

13. F

or p

erso

nal u

se o

nly.

Page 4: Virginia M-Y Lee,1 Michel Goedert,2 and John Q Trojanowski

P1: FUM

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

1124 LEE ¥ GOEDERT ¥ TROJANOWSKI

Figure 1 Schematic representation of the human tau gene and the six central nervous system(CNS) tau isoforms generated by alternative mRNA splicing. The human tau gene contains 16exons, 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 thehuman CNS. E4a (striped box), which is also not transcribed in the human CNS, is expressed inthe peripheral nervous system, leading to the larger tau isoforms, termed big tau (see text). Theblack 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 tauprotein have been discovered and extensively characterized (for review, see Bu´eeet al 2000). Most notably, tau binds to and stabilizes microtubules (MTs), in ad-dition to promoting MT polymerization (Weingarten et al 1975, Cleveland et al1977b). The MT binding domains of tau are localized to the carboxy-terminal halfof the molecule within the four MT binding motifs. These motifs are composed ofhighly conserved 18–amino acid long binding elements separated by flexible, butless conserved, interrepeat sequences that are 13–14 amino acids long (Himmleret al 1989, Lee et al 1989, Butner & Kirschner 1991). The binding of tau to MTsis a complex process mediated in part by a flexible array of weak MT binding sitesthat are distributed throughout the MT binding domain delineated by these repeatsand their interrepeat sequences (Lee et al 1989, Butner & Kirschner 1991). In ad-dition, sequences flanking the repeats contribute to microtubule binding (Gustkeet al 1994). 4R-tau isoforms are more efficient at promoting MT assembly andhave a greater MT binding affinity than do 3R-tau isoforms (Goedert & Jakes1990, Butner & Kirschner 1991). It is interesting to note that the interrepeat se-quence between the first and second MT binding repeats has more than twicethe binding affinity of any individual MT binding repeat (Goode & Feinstein1994). This region is unique to 4R-tau and is believed to be responsible for thehigher MT binding affinity of 4R-tau isoforms compared with 3R-tau isoforms(Goedert & Jakes 1990). Notably, because other microtubule-associated proteins

Ann

u. R

ev. N

euro

sci.

2001

.24:

1121

-115

9. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by O

pen

Uni

vers

ity o

n 07

/11/

13. F

or p

erso

nal u

se o

nly.

Page 5: Virginia M-Y Lee,1 Michel Goedert,2 and John Q Trojanowski

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 didnot have any overt phenotype (Harada et al 1994). However, a more recent studyhas reported some subtle behavioral impairments in older knockout mice (Ikegamiet al 2000).

TAU PHOSPHORYLATION AND REGULATIONOF TAU FUNCTIONS

There are 79 potential serine (Ser) and threonine (Thr) phosphate acceptor residuesin the longest tau isoform, and phosphorylation at∼30 of these sites has beenreported in normal tau proteins (reviewed in Billingsley & Kincaid 1997, Bu´eeet al 2000). Tau phosphorylation is developmentally regulated such that fetal tau ismore highly phosphorylated in embryonic compared with adult CNS (Kanemaruet al 1992, Bramblett et al 1993, Goedert et al 1993, Watanabe et al 1993), andthe degree of phosphorylation of all six tau isoforms decreases with age, probablybecause of the activation of phosphatases (Mawal-Dewan et al 1994). The tauphosphorylation sites are clustered in regions flanking the MT binding repeats,and it is well established that increasing tau phosphorylation negatively regulatesMT 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 MTbinding 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 rolein reducing the binding of tau to MTs (Biernat et al 1993). A similar role may beconsidered for phosphorylation of Ser396, which is located adjacent to the carboxy-terminal end of the fourth MT binding domain (Bramblett et al 1993), but otherdata argue that phosphorylation of neither of these sites is sufficient to eliminatethe binding of tau to MTs (Seubert et al 1995). It is interesting that both sites arephosphorylated in fetal tau and that they are hyperphosphorylated in all six tauisoforms that form abnormal paired helical filaments (PHFs) in the neurofibrillarytangles (NFTs) of the AD brain (Seubert et al 1995). The regulation of the bindingof 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 aheptapeptide sequence (224KKVAVVR 230) located within a proline-rich domain,amino-terminal to the MT binding domains, promotes MT binding in combinationwith the repeat regions (Goode et al 1997). Thus, it is likely that phosphorylation atmultiple sites, especially those flanking the MT binding repeats, but also additionalintra- and intermolecular interactions, regulate the MT binding function of tau.Only little is known about the regulation of tau phosphorylation. One study hasreported increased phosphorylation of tau at Ser202/Thr205 in mice that lackReelin or both the very-low-density lipoprotein receptor and the apolipoproteinE receptor 2 (Hiesberger et al 1999). It suggests that tyrosine phsophorylationof the disabled-1 adaptor protein may play a role in regulating the level of tau

Ann

u. R

ev. N

euro

sci.

2001

.24:

1121

-115

9. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by O

pen

Uni

vers

ity o

n 07

/11/

13. F

or p

erso

nal u

se o

nly.

Page 6: Virginia M-Y Lee,1 Michel Goedert,2 and John Q Trojanowski

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 toadditional phosphorylation sites in tau and to mice mutant for other componentsof this developmental pathway.

For the reasons discussed above, considerable research has focused on theprotein kinases and protein phosphatases that regulate tau phosphorylation, and alarge 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, Buee et al 2000, Hong et al 2000). These kinases include mito-gen-activated protein kinase (Drewes et al 1992, Drechsel et al 1992, Goedertet al 1992a), glycogen synthase kinase 3β (GSK-3β) (Hanger et al 1992,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).In addition, several members of the family of stress-activated protein kinases alsophosphorylate 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 onlyin vitro evidence to implicate specific kinases and that it remains unclear what rolethey play in the in vivo phosphorylation of tau.

Recent data have implicated two protein kinases, GSK-3β and cdk5, in the invivo regulation of tau phosphorylation. GSK-3β is a Ser/Thr kinase that is abundantin brain and associates with MTs (Mandelkow et al 1992, Ishiguro et al 1994, Singhet al 1995, Takahashi et al 1995, Cohen 1999). Cotransfection of nonneuronalcells with human tau and GSK-3β induces hyperphosphorylation of tau associatedwith 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 aphosphatidylinositol 3-kinase and protein kinase B–dependent signaling pathway(Hong & Lee 1997). In addition, direct inhibition of GSK-3β by lithium salts orATP competitive inhibitors reduces tau phosphorylation and affects MT stability(Hong et al 1997, Munoz-Montano et al 1997, Lovestone et al 1999, Takahashiet al 1999, Leost et al 2000). Cdk5 is a Ser/Thr protein kinase highly enriched inneurons that colocalizes to the cytoskeleton and contributes to the phosphorylationof tau (Baumann et al 1993, Kobayashi et al 1993, Lew & Wang 1994). It isactivated by interaction with regulatory subunits, the best characterized of whichis 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 onthe 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 neededto determine the relative contributions of individual kinases to tau phosphorylationin vivo.

Because protein phosphatases are required for counterbalancing the effects oftau protein kinases, they have also been an intense focus of research. A numberof studies have implicated several phosphatases in regulating tau phosphorylation,including PP1, PP2A, PP2B, and PP2C (reviewed in Billingsley & Kincaid 1997,

Ann

u. R

ev. N

euro

sci.

2001

.24:

1121

-115

9. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by O

pen

Uni

vers

ity o

n 07

/11/

13. F

or p

erso

nal u

se o

nly.

Page 7: Virginia M-Y Lee,1 Michel Goedert,2 and John Q Trojanowski

P1: FUM

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

NEW INSIGHTS INTO TAUOPATHIES 1127

Buee 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 inhuman brain tissue, and they dephosphorylate tau in a site-specific manner. Bothenzymes 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 themajor activity toward tau phosphorylated by a number of protein kinases (Goedertet al 1992a, 1995a). PP1 and PP2A bind to tau, and this interaction is believedto mediate an association with MTs (Sontag et al 1995, Liao et al 1998). PP2Ahas also been demonstrated to bind directly to MTs, an interaction that regulatesits activity in vitro (Sontag et al 1999). Lastly, inhibition of PP1 and PP2A byokadaic acid in cultured human NT2N neurons results in increased tau phospho-rylation, accompanied by decreased tau binding to MTs, selective destruction ofstable MTs, and rapid degeneration of axons (Merrick et al 1997). As with thekinases implicated in the phosphorylation of tau, further studies are necessary todefine the specific role of individual phosphatases in the in vivo regulation of thephosphorylation state of tau.

AD NEUROFIBRILLARY PATHOLOGY IS MADE OFABNORMALLY PHOSPHORYLATED TAU

Filamentous neuronal or neuronal and glial tau inclusions associated with the de-generation of affected brain regions are the defining neuropathological features oftauopathies. In AD, NFTs and neuropil thread pathology are found in conjunctionwith the deposition ofβ-amyloid (Aβ) fibrils in the extracellular space. By lightmicroscopy, the neurofibrillary lesions of AD are stained with anti-tau antibod-ies (Brion et al 1985, Grundke-Iqbal et al 1986). Ultrastructurally, the dominantcomponents of neurofibrillary lesions in AD are paired helical filaments (PHFs)and straight filaments (Kidd 1963). PHFs are composed of two strands of filamenttwisted around one another with a periodicity of 80 nm and a width varying from8 to 20 nm (Crowther & Wischik 1985) whereas straight filaments lack this helicalperiodicity (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). Analysisof PHFs purified from AD brains by sodium dodecyl sulphate–polyacrylamidegel electrophoresis has revealed three major bands of approximately 68, 64, and60 kDa, as well as a minor band of approximately 72 kDa (Greenberg & Davies1990, Lee et al 1991) (Figure 2). Upon dephosphorylation, six bands are resolvedthat correspond to the six isoforms of tau found in adult human brain (Lee et al1991, Greenberg et al 1992, Goedert et al 1992b). The relative proportions ofthe 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 (seeTrojanowski & Lee 1994, Morishima-Kawashima et al 1995, Goedert et al 1995b,Hong et al 1998). Although many phosphorylation sites identified in PHFtau

Ann

u. R

ev. N

euro

sci.

2001

.24:

1121

-115

9. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by O

pen

Uni

vers

ity o

n 07

/11/

13. F

or p

erso

nal u

se o

nly.

Page 8: Virginia M-Y Lee,1 Michel Goedert,2 and John Q Trojanowski

P1: FUM

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

1128 LEE ¥ GOEDERT ¥ TROJANOWSKI

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 withphosphorylation-independent anti-tau antibodies. Nondephosphorylated insoluble tau from thebrain 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 andR406W), runs as three major bands of 68, 64, and 60 kDa and as a minor, variable band of 72kDa. 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 withthe P301L mutation, the two prominent 68- and 64-kDa insoluble tau bands are detected (the72-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 isselective 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 isdeposited 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 isoformsare expressed in the soluble fraction in PiD. Major tau proteins are depicted by solid bars and thethickness of the bars correlates with the relative abundance of the specific tau isoform. A dashedbar 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 isolatedfrom biopsies of normal human brain (Matsuo et al 1994), it is clear that PHFtauis hyperphosphorylated and abnormally phosphorylated (Morishima-Kawashimaet al 1995, Hasegawa et al 1996, Hoffmann et al 1997, Zheng-Fischhofer et al1998).

Ann

u. R

ev. N

euro

sci.

2001

.24:

1121

-115

9. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by O

pen

Uni

vers

ity o

n 07

/11/

13. F

or p

erso

nal u

se o

nly.

Page 9: Virginia M-Y Lee,1 Michel Goedert,2 and John Q Trojanowski

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 inthe dysregulation of tau phosphorylation in the AD brain (for detailed reviews, seeBillingsley & Kincaid 1997, Buee et al 2000, Hong et al 2000). A recent study hassuggested 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 brainsof AD patients. The calcium-dependent cysteine protease calpain is believed togenerate 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 thebinding of p25 to cdk5 constitutively activated cdk5. Finally, the expression of thecdk5/p25 complex in various cell lines increased tau phosphorylation and disruptedthe cytoskeletal network. Thus, it is possible that the cdk5/p25 complex may playa mechanistic role in the conversion of normal tau into PHFtau in AD; however, itwill be important to confirm and extend these results in additional in vitro and invivo studies.

The mechanisms underlying PHF formation in neurons are still unclear, but itis possible that hyperphosphorylation disengages tau from MTs, thereby increas-ing the pool of unbound tau. Unbound tau may be more resistant to degradationand more prone to aggregate than MT-bound tau. This suggests that an increasedability of pathological tau to intereact with MTs may be beneficial. The organicosmolytes trimethylamine N-oxide and betaine have been shown to increasetau-promoted assembly of MTs (Tseng & Graves 1998). These compounds werealso shown to restore the ability of tau phosphorylated by cAMP-dependent proteinkinase or GSK-3β to promote MT assembly (Tseng et al 1999). Tau is a highlyflexible, 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 carryingFTDP-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 MTsgenerates some ordered structures, indicating that MTs can induce conformationalchanges in tau (Woody et al 1983). Trimethylamine N-oxide and betaine probablyinduce a tubulin and/or tau conformational change that favors assembly.

Along related lines, Lu et al (1999) demonstrated that the prolyl isomerase Pin1binds to a single phosphothreonine residue (Thr231) in tau and that this restoredthe ability of tau phosphorylated by cdc2 kinase to intereact with MTs. Pin1 wasreported to copurify with PHFs, resulting in a depletion of soluble Pin1 in ADbrain. Because depletion of Pin1 is believed to induce mitotic arrest and apoptoticcell death, its sequestration in NFTs could contribute to neurodegeneration.

SYNTHETIC TAU FILAMENTS

Hyperphosphorylation is believed to be an early event in the pathway that leadsfrom soluble to insoluble and filamentous tau protein (Braak et al 1994). However,it is unclear whether it is sufficient for assembly into filaments. Currently, thereis no experimental evidence that links hyperphosphorylation of tau to filament

Ann

u. R

ev. N

euro

sci.

2001

.24:

1121

-115

9. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by O

pen

Uni

vers

ity o

n 07

/11/

13. F

or p

erso

nal u

se o

nly.

Page 10: Virginia M-Y Lee,1 Michel Goedert,2 and John Q Trojanowski

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 isoformsand the ease with which tau fragments can be expressed has facilitated studiesaimed at producing synthetic tau filaments. Early experiments had shown thatPHF-like filaments can be assembled in vitro from bacterially expressed, non-phosphorylated 3R fragments of tau (Crowther et al 1992, Wille et al 1992). Theformation of these filaments lent strong support to the view that the repeat regionof tau is the only component necessary for the morphological appearance of thePHF. However, these studies failed to provide any insight into filament formationin vivo because the tau filaments were obtained only with truncated tau under non-physiological conditions. This contrasts with PHFs from AD brain, which consistof full-length tau protein (Goedert et al 1992b).

More recently, experiments using sulphated glycosaminoglycans (GAGs) tostimulate 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(Goedert et al 1996, Perez et al 1996). Assembly of individual 3R-tau isoformsgave filaments with a typical paired-helical-like morphology, when incubated withheparin or heparan sulphate, whereas assembly of individual 4R-tau isoform gavefilaments with a straight appearance. By immunoelectron microscopy, the paired-helical-like filaments were decorated by antibodies directed against the aminoand 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-inducedfilament assembly (von Bergen et al 2000).

Assembly of tau into filaments in the presence of sulphated GAGs occurs aftera 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 al1996). However, it has been reported that phosphorylation at other sites, such asSer214 and Ser262, is strongly inhibitory toward assembly (Schneider et al 1999).Subsequent to this work, RNA (Kampers et al 1996, Hasegawa et al 1997) andarachidonic acid (Wilson & Binder 1997) were also shown to induce the bulkassembly of full-length recombinant tau into filaments. This work has providedrobust methods for the assembly of full-length tau into filaments. Pathologicalcolocalization of sulphated GAGs (Snow et al 1990, Goedert et al 1996, Verbeeket al 1999) and RNA (Ginsberg et al 1998) with hyperphosphorylated tau proteinsuggests 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 thedeposition and fibrillization of Aβ peptides to form extracellular senile plaques isthe central event that causes formation of NFTs and neuronal loss (Hardy & Allsop

Ann

u. R

ev. N

euro

sci.

2001

.24:

1121

-115

9. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by O

pen

Uni

vers

ity o

n 07

/11/

13. F

or p

erso

nal u

se o

nly.

Page 11: Virginia M-Y Lee,1 Michel Goedert,2 and John Q Trojanowski

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 thanAD, which share an abundant filamentous tau pathology and brain degeneration inthe absence of extracellular amyloid deposits (Table 1). Progressive supranuclearpalsy (PSP), corticobasal degeneration (CBD), and Pick’s disease (PiD) are threesuch disorders. A recent consensus conference has classified them as disordersbelonging to a group of diseases known as frontotemporal dementia (FTD) (Pick’sConference 2001). Clinically, PSP is characterized by supranuclear gaze palsy aswell as by prominent postural instability (Steele et al 1964). Neuropathologically,PSP is characterized by atrophy of the basal ganglia, subthalamus, and brainstem,with corresponding neuronal loss and gliosis. Within these brain regions, there isa high density of fibrillary tau pathology, including neuropil threads, and NFTs thatare typically round or globose (Pollock et al 1986, Hauw et al 1994, Litvan et al1996). Glial fibrillary tangles in both astrocytes (tufted astrocytes) and oligoden-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 neurofibrillarylesions has revealed 15- to 18-nm straight filaments, and filaments with a longperiodicity have also been observed (Tellez-Nagel & Wisniewski 1973, Roy et al1974).

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 highMr bands(68 and 64 kDa) are present (the minor 72-kDa band is variably detected) (Flamentet 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 al1999). They exhibit the same profile of phosphorylation-dependent tau epitopesas 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 tobe increased in the brainstem but not in the cortex, which is consistent with thedistribution of the neurofibrillary pathology (Chambers et al 1999).

Polymorphisms in thetau gene may contribute to the risk of developing PSPbecause a polymorphic dinucleotide repeat in the intron between E9 and E10 of thetaugene has been linked to PSP (Conrad et al 1997). Subjects with the homozygoustau allele A0, characterized by 11 TG repeats, were found to be overrepresented inPSP 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, Morriset al 1999a). Moreover, two extendedtau gene haplotypes consisting of eightcommon single-nucleotide polymorphisms in addition to the dinucleotide repeatpolymorphism have been described (Baker et al 1999). The haplotypes are incomplete linkage dysequilibrium and span the entire humantau gene. The morecommon haplotype, H1, is significantly overrepresented in Caucasians with PSP.In addition, two missense mutations in E4a are associated with the H1 haplotypeand have been linked to PSP, and a 238-bp deletion in the intron flanking E10 of

Ann

u. R

ev. N

euro

sci.

2001

.24:

1121

-115

9. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by O

pen

Uni

vers

ity o

n 07

/11/

13. F

or p

erso

nal u

se o

nly.

Page 12: Virginia M-Y Lee,1 Michel Goedert,2 and John Q Trojanowski

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 andthus 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 haplotypeand the A0 allele to the pathogenesis of PSP are unknown, but it is possible thatthe 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 thecerebral cortex, deep cerebellar nuclei, and substantia nigra, in association withprominent neuronal achromasia (Rebeiz et al 1967, 1968). Neuropathological ex-amination shows depigmentation of the substantia nigra, as well as an asymmetricfrontoparietal atrophy that is often most severe in the pre- and postcentral regions.In affected regions, there is neuronal loss with spongiosis, gliosis, and prominentglial and neuronal intracytoplasmic filamentous tau pathology (Iwatsubo et al 1994,Mori et al 1994). The glial tau pathology in CBD consists of characteristic astro-cytic plaques (Feany & Dickson 1995), as well as numerous tau-immunoreactiveinclusions 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 extensiveaccumulation of tau-immunoreactive neuropil threads throughout gray and whitematter (Feany & Dickson 1995, Feany et al 1996). The tau filaments in CBDinclude 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 inthat it consists of two major bands of 64 and 68 kDa and a variable, minor bandof 72 kDa (Ksiezak-Reding et al 1994, Bu´ee-Scherrer et al 1996) (Figure 2).However, the isoforms present in the tau pathology of CBD may differ from thosefound in PSP. Antibodies specific for the insert encoded by E3 did not detect thefibrillary tau pathology in CBD either biochemically or immunohistochemically instudies 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 studydemonstrated that the fibrillary inclusions in CBD are composed predominantlyof 4R-tau isoforms that also contain the inserts encoded by E2 and to E3. Anothersimilarity between PSP and CBD was recently described by Di Maria et al (2000),who showed that CBD is associated with the A0 allele of thetau gene, as well asthe H1 haplotype. Thus, the current biochemical and genetic data strongly suggestthat there is a substantial overlap between PSP and CBD. This is also apparentwith respect to the clinical (Hauw et al 1994) and pathological (Feany et al 1996)features. Rather than representing two separate and distinct disorders, PSP andCBD may be different phenotypic manifestations of the same underlying diseaseprocess.

PiD, a variant of FTD, is defined neuropathologically by the presence of tau-immunoreactive Pick bodies (Constantinidis et al 1974, Pollock et al 1986, Feanyet al 1996, Pick’s Conference 2001). Neuropathologically, it is characterizedby a frontotemporal lobar and limbic atrophy associated with marked neuronalloss, spongiosis, and gliosis, with ballooned neurons and Pick bodies (Lund &

Ann

u. R

ev. N

euro

sci.

2001

.24:

1121

-115

9. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by O

pen

Uni

vers

ity o

n 07

/11/

13. F

or p

erso

nal u

se o

nly.

Page 13: Virginia M-Y Lee,1 Michel Goedert,2 and John Q Trojanowski

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 antibodiesto 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 ofwide, 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 distinctfrom that in AD, CBD, and PSP in that it comprises two major bands of 60 and 64kDa and a variable, minor band of 68 kDa (Bu´ee-Scherrer et al 1996, Delacourteet al 1996, Lieberman et al 1998) (Figure 2). Because the two major PiD taubands appear to lack the MT binding repeat encoded by E10, they are believedto 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 detecteda signal in Pick bodies and PiD tau using an antibody specific for phosphorylatedSer262 (Lieberman et al 1998). This may reflect heterogeneity of phosphorylationat 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-specificdiagnostic 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 appropriatenomenclature for this form of FTD (Mann 1998). However, this may changesoon because one study has defined this neuropathology more precisely usingquantitative morphometric methods (Arnold et al 2000). Moreover, recent evi-dence suggests that FTLD (DLDH) may be a novel tauopathy that is caused bya selective reduction or complete loss of all six brain tau isoforms in affectedand unaffected brain regions (Zhukareva et al 2001). The explanation for this isenigmatic because there was no concomitant loss of tau mRNA compared withcontrol and AD brains. Although the majority of FTD patients with FTLD (DLDH)neuropathology showed a dramatic loss of tau protein in brain regions with andwithout neuronal degeneration, others showed less-substantial but still statisticallysignificant reductions in brain tau levels. Although the pathogenic mechanism un-derlying this marked reduction in all six brain tau proteins in FTLD (DLDH) is notknown, the consequence of this loss may be similar to the losses of tau functionresulting from some of thetaugene mutations in FTDP-17.

FAMILIAL TAUOPATHIES—FTDP-17 SYNDROMES

The group of syndromes known as FTDP-17 consists of autosomal-dominantlyinherited neurodegenerative diseases with diverse, but overlapping, clinical andneuropathological features (Foster et al 1997). Neuropathologically, they all showthe presence of an abundant filamentous tau pathology in nerve cells, and for some

Ann

u. R

ev. N

euro

sci.

2001

.24:

1121

-115

9. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by O

pen

Uni

vers

ity o

n 07

/11/

13. F

or p

erso

nal u

se o

nly.

Page 14: Virginia M-Y Lee,1 Michel Goedert,2 and John Q Trojanowski

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, Honget al 2000). The first such disorder was linked to chromosome 17 in 1994, whenWilhelmsen et al (1994) described a familial disease they called “disinhibition-dementia-parkinsonism-amyotrophy complex” and demonstrated genetic linkageof this disease to chromosome 17q21-22. Subsequently, a number of related neu-rodegenerative disorders were linked to the same region on chromosome 17 (Wijkeret al 1996, Bird et al 1997, Foster et al 1997, Heutink et al 1997, Murrell et al1997, Lendon et al 1998). Clinically, they are characterized primarily by FTD andparkinsonism (Foster et al 1997), but the different FTDP-17 syndromes appear toreflect the burden of tau pathology and degeneration in brain regions known tosubserve specific cognitive, executive, or motor functions. Despite this pheno-typic heterogeneity, the neuropathology of FTDP-17 is characterized by markedneuronal loss in affected brain regions, with extensive neuronal or neuronal andglial fibrillary pathology composed of hyperphosphorylated tau protein, but with-out evidence of Aβ deposits or other disease-specific brain lesions in the majorityof the cases (Murrell et al 1999, Lippa et al 2000, Rizzini et al 2000, Spillantiniet al 2000).

Because thetau gene had been localized to chromosome 17q21-22, it wasan obvious candidate for the disease locus. In 1998, several groups identifiedpathogenic mutations in thetau gene that segregated with FTDP-17 (Clark et al1998, Dumanchin et al 1998, Hutton et al 1998, Poorkaj et al 1998, Spillantiniet al 1998c). To date,>20 distinct pathogenic mutations in thetau gene havebeen identified in a large number of families with FTDP-17 (Table 2, Figure 3).Eleven missense mutations in coding regions of thetau 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 al2000), P301L (Clark et al 1998, Dumanchin et al 1998, Hutton et al 1998, Birdet 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 al1999), N296N (Spillantini et al 2000), and S305S (Stanford et al 2000)] as wellas a deletion mutation [1K280 (Rizzu et al 1999)] have also been identified. Inaddition, five substitutions in six different positions of the intron following E10have 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, Huttonet 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, additionalpathogenic mutations may be present in other introns of thetau gene. Thus, amutation in the intron following E9 has been described in a patient with familialFTD (Rizzu et al 1999). It disrupts one of the several (A/T)GGG repeats that mayplay a role in the regulation of the alternative splicing of E10.

Ann

u. R

ev. N

euro

sci.

2001

.24:

1121

-115

9. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by O

pen

Uni

vers

ity o

n 07

/11/

13. F

or p

erso

nal u

se o

nly.

Page 15: Virginia M-Y Lee,1 Michel Goedert,2 and John Q Trojanowski

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, personalcommunication

G272V E9, R1 No change Reduced FTDP-17 Hutton et al (1998)

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)

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-likeCBD-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)

aFTDP-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.

Ann

u. R

ev. N

euro

sci.

2001

.24:

1121

-115

9. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by O

pen

Uni

vers

ity o

n 07

/11/

13. F

or p

erso

nal u

se o

nly.

Page 16: Virginia M-Y Lee,1 Michel Goedert,2 and John Q Trojanowski

P1: FUM

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

1136 LEE ¥ GOEDERT ¥ TROJANOWSKI

Figure 3 Schematic representation of mutations in the tau gene identified in frontotemporaldementia and parkinsonism linked to chromosome 17. The longest human brain tau isoform isshown with known coding region mutations indicated above. The gray boxes near the aminoterminus represent the alternatively spliced inserts encoded by exons (E)2 and E3, whereas theblack 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 andthe 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 supportin favor of the hypothesis that FTDP-17 mutations lead to tau dysfunction anddisease by one or more of three distinct mechanisms. Intronic and some exonicmutations affect the alternative splicing of E10 and consequently alter the relativeproportion of 4R-tau and 3R-tau. The other exonic mutations impair the abilityof tau to bind MTs and to promote MT assembly. Some of these mutations alsopromote the assembly of tau into filaments. Moreover, additional mechanismsmay 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 5′ splice site ofE10, 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 ofthese mutations, there is a relative increase in E10-containing tau mRNAs, and thisprobably reflects increased utilization of the E10 5′ splice site, as demonstrated inexon trapping experiments. Biochemical analysis of insoluble tau extracted fromautopsied FTDP-17 brain tissue of patients with these mutations reveals exclusively4R-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 unaffectedregions of FTDP-17 brains (Hong et al 1998, Spillantini et al 1998c, Goedert et al1999b, Yasuda et al 2000).

Ann

u. R

ev. N

euro

sci.

2001

.24:

1121

-115

9. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by O

pen

Uni

vers

ity o

n 07

/11/

13. F

or p

erso

nal u

se o

nly.

Page 17: Virginia M-Y Lee,1 Michel Goedert,2 and John Q Trojanowski

P1: FUM

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

NEW INSIGHTS INTO TAUOPATHIES 1137

The regulation of splicing of E10 in thetaugene appears to be complex and mayinvolve multiplecis-acting regulatory elements that either enhance or inhibit theutilization of the E10 5′ splice site, many of which are affected by the mutationsidentified in thetau gene (D’Souza et al 1999, Grover et al 1999, D’Souza &Schellenberg 2000, Gao et al 2000, Jiang et al 2000). Splicing regulatory elementswithin E10 appear to include an exon-splicing enhancer (ESE) and an exon splicingsilencer (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-richsequence, and an AC-rich sequence (D’Souza & Schellenberg 2000). Immediatelydownstream of the ESE within E10 is a purine-rich ESS. The flanking exons of thetaugene also appear to affect E10 splicing (Gao et al 2000). For example, it appearsthat E9 and E11 exert opposite effects, i.e. E9 may promote E10 splicing, whereasE11 may suppress it. Lastly, intronic sequences immediately downstream of E10inhibit its splicing (D’Souza et al 1999, Grover et al 1999, D’Souza & Schellenberg2000, Gao et al 2000, Jiang et al 2000). This inhibition may be secondary to theformation of a stem-loop structure that sequesters the E10 5′ splice site fromthe 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 thethree-dimensional structure of a 25-nucleotide-long RNA from the E10 5′-intronjunction by nuclear magnetic resonance spectroscopy has shown that this sequenceforms a stable, folded stem-loop structure (Varani et al 1999, 2000). The stemconsists of a single G-C base pair that is separated from a double helix of 6 bp byan unpaired adenine (Figure 3). As is often the case with single-nucleotide purinebulges, the unpaired adenine at position−2 does not extrude into solution butintercalates into the double helix. The apical loop consists of six nucleotides thatadopt multiple conformation in rapid exchange. Known intronic mutations andthe mutations in codon 305 are located in the upper part of the stem and reducethe thermodynamic stability of the stem loop (Varani et al 1999, 2000; Yasudaet al 2000). Moreover, the relative proportions of 3R-tau and 4R-tau isoformsfrom nonhuman species correlate with the predicted stability of this stem-loopstructure (Grover et al 1999). However, another study has concluded that this ESSmay function as a linear sequence that is independent of a stem-loop structure(D’Souza & Schellenberg 2000).

Pathogenic FTDP-17 mutations in thetau gene may alter E10 splicing by af-fecting several of the regulatory elements described above. Thus, the intronicmutations, 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 al1999) (Figure 3). The S305N mutation and the+3 intronic mutation may alsoenhance E10 splicing by increasing the strength of the 5′ splice site (Spillantiniet al 1998c, Iijima et al 1999). However, the finding that the S305S mutation thatweakens the E10 5′ splice site also leads to a predominance of 4R-tau argues againstthis effect of these mutations (Stanford et al 2000). The N279K mutation mayimprove the function of the ESE by lengthening the purine-rich sequence withinthis regulatory element (TAAGAA to GAAGAA), thus enhancing E10 splicing

Ann

u. R

ev. N

euro

sci.

2001

.24:

1121

-115

9. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by O

pen

Uni

vers

ity o

n 07

/11/

13. F

or p

erso

nal u

se o

nly.

Page 18: Virginia M-Y Lee,1 Michel Goedert,2 and John Q Trojanowski

P1: FUM

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

1138 LEE ¥ GOEDERT ¥ TROJANOWSKI

(D’Souza & Schellenberg 2000). Moreover, the thymidine nucleotide present inthe wild-type (WT) sequence may function as an inhibitor of splicing (Tanaka et al1994). The observation that the1K280 mutation, which deletes the three adjacentpurine residues (AAG), abolishes E10 splicing supports this hypothesis (D’Souzaet al 1999). The silent L284L mutation that enhances E10 splicing may do soby disrupting a potential ESS (UUAG to UCAG) (Si et al 1998, D’Souza et al1999). However, because mutation of this consensus sequence does not increaseE10 splicing (D’Souza & Schellenberg 2000), a second possibility is that the mu-tation lengthens the AC-rich element within the ESE. Thus, the L284L mutationmay affect either an enhancing or an inhibiting regulatory splicing element. Theeffect of the N296N mutation on splicing of E10 is probably due to disruption ofan 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-tauand 3R-tau may bind to distinct sites on MTs (Goode & Feinstein 1994, Goodeet al 1997), and it is possible that a specific ratio of tau isoforms is necessary fornormal MT function. Thus, the altered ratio of 3R/4R-tau may directly affect MTfunction. In addition, overproduction of 4R-tau may lead to an excess of free tauin the cytoplasm, leading to its hyperphosphorylation and assembly into filaments.

In contrast to the FTDP-17 mutations discussed above, other mutations alter theability of tau to interact with MTs. Specifically, mutations K257T, G272V,1K280,P301L, P301S, V337M, G389R, and R406W reduce the binding of tau to MTsand decrease its ability to promote MT assembly in in vitro assays (Hasegawaet al 1998, Hong et al 1998, Bugiani et al 1999, D’Souza et al 1999, Murrellet 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 al1999). Similar effects on MT function are observed when tau is expressed in avariety of cell lines, including SHSY5Y neuroblastoma cells (Dayanandan et al1999), Chinese hamster ovary (CHO) cells (Dayanandan et al 1999, Matsumuraet al 1999, Vogelsberg-Ragaglia et al 2000), monkey kidney (COS) cells (Arawakaet al 1999, Sahara et al 2000), and Sf9 insect cells (Frappier et al 1999). Expressionof 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 bindingand/or function, both in in vitro assays and in transfected cell lines (DeTure et al2000, Sahara et al 2000). The discrepancies between these and other studies aremost likely due to the differences in the levels of expression, the methods used forthe quantification of tau levels, and the binding of tau to MTs. Nevertheless, evenif these missense mutations cause only a modest reduction in MT binding affinity,this could have large cumulative effects on affected neurons over the human lifespan. Furthermore, increased cytosolic concentrations of unbound mutant tau pro-teins may facilitate aggregation of these abnormal proteins, with or without theirWT counterparts, into filamentous inclusions.

Ann

u. R

ev. N

euro

sci.

2001

.24:

1121

-115

9. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by O

pen

Uni

vers

ity o

n 07

/11/

13. F

or p

erso

nal u

se o

nly.

Page 19: Virginia M-Y Lee,1 Michel Goedert,2 and John Q Trojanowski

P1: FUM

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

NEW INSIGHTS INTO TAUOPATHIES 1139

A subset of missensetau gene mutations may cause FTDP-17, at least in part,by promoting tau aggregation. Several studies have demonstrated that some ofthese mutations, including K257T, G272V,1280K, P301L, P301S, V337M, andR406W, promote heparin- or arachidonic acid–induced tau filament formation invitro relative to WT tau (Arrasate et al 1999, Nacharaju et al 1999, Goedert et al1999a, 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 the1K280 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 the1K280,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-Ragagliaet al 2000).

All known mutations in thetau gene lead to the formation of filaments madeof hyperphosphorylated tau protein (Crowther & Goedert 2000). However, thefibrillary lesions observed with mutations in E10 or the intron following E10 arebiochemically and ultrastructurally distinct from the lesions caused by mutationsthat are located outside E10. Coding region mutations located outside E10 affect allsix isoforms of tau. Thus, as one might predict, tau fibrillary lesions are composedof all six tau isoforms (Hong et al 1998, Murrell et al 1999, Van Swieten et al1999) (Figure 2). For some mutations (V337M and R406W), the morphologiesand biochemical characteristics of tau filaments are indistinguishable from thoseof AD (Spillantini et al 1996, Hong et al 1998, Van Swieten et al 1999). Othercoding region mutations located outside E10 (K252T, G272V, E342V, and G389R)give rise to a tau pathology that closely resembles that of PiD (Spillantini et al1998b, 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 aggregationof 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 filamentsobserved in 4R-tau disorders, particularly CBD (Reed et al 1998, Spillantini et al1998b, 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 disinhibitiondementia 2 (HDDD2), which appears similar to some of the syndromes seen inFTDP-17 kindreds, has been reported to show linkage to 17q21-22 with a lod(logarithm of odds) score of 3.68; however, notau gene mutation or any othergenetic abnormality has been identified in this family (Lendon et al 1998). It issurprising, however, that recent studies of three brains from affected members ofthe HDDD2 kindred revealed that HDDD2 shares significant neuropathologicaland biochemical abnormalities with sporadic FTD patients classified as FTLD(DLDH) (Lendon et al 1998, Zhukareva et al 2001). As discussed above regardingFTLD (DLDH), the loss of tau proteins in several of the HDDD2 brains, together

Ann

u. R

ev. N

euro

sci.

2001

.24:

1121

-115

9. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by O

pen

Uni

vers

ity o

n 07

/11/

13. F

or p

erso

nal u

se o

nly.

Page 20: Virginia M-Y Lee,1 Michel Goedert,2 and John Q Trojanowski

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 maybe controlled posttranscriptionally, either at the level of tau mRNA translation orthrough mechanisms that regulate mRNA stability. Thus, the HDDD2 kindredappears to be the familial counterpart of sporadic FTLD (DLDH), both of whichmay define a novel and distinct tauopathy caused by a reduction of brain tau.

Although the biochemical and structural characteristics of the tau aggregatesin 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 phenotypesand topographical distributions of pathology is more enigmatic. For example, itis not clear why the clinical and neuropathologic phenotype of individuals withFTDP-17 mutations ranges from FTD (including subtypes thereof, such as PiD,CBD, and PSP) to multisystem neurodegeneration. However, sometaugene mu-tations cause a similar phenotype in different families or in different members ofthe same family. For instance, the N279K mutation typically causes a phenotypereminiscent of PSP with superimposed dementia (Reed et al 1998, Delisle et al1999, Yasuda et al 1999). In contrast, there are several clinical and pathologicdescriptions of families with the P301L mutation that demonstrate a highly vari-able phenotype ranging from PSP to CBD to PiD (Spillantini et al 1998b, Birdet al 1999, Mirra et al 1999, Nasreddine et al 1999). Even more perplexing is thereport of two brothers from one P301L family (Bird et al 1999), with frontal lobedegeneration in one individual and PSP-like pathology in the other. Similarly, ina family with the P301S mutation, one individual presented with FTD whereashis son presented clinically with CBD (Bugiani et al 1999). Although only a fewreports 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 pathologicdistinctions between them may be due to other genetic and/or epigenetic factorsthat modify the effects of the primary mutation. Currently, the specific geneticand/or environmental modifiers that might determine the phenotype of a specificindividual remain unknown, but these are fields of active investigation, and thegeneration of animal models of tau-mediated neurodegeneration may facilitatethis 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 ofa variety of neurodegenerative disorders. In addition, they may be useful modelsfor the development and testing of novel therapies. Early efforts to produce an-imal models with tau pathology were largely based on the hypothesis that thedevelopment 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 al1996, Sturchler-Pierrat et al 1997). More recently, several animal models of taupathology were produced by overexpressing human tau proteins (Table 3).

Ann

u. R

ev. N

euro

sci.

2001

.24:

1121

-115

9. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by O

pen

Uni

vers

ity o

n 07

/11/

13. F

or p

erso

nal u

se o

nly.

Page 21: Virginia M-Y Lee,1 Michel Goedert,2 and John Q Trojanowski

P1: FUM

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

NEW INSIGHTS INTO TAUOPATHIES 1141

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

Initial reports described TG mouse lines expressing 4R2N or 3R0N human tauutilizing cDNA constructs with either the Thy1 or the 3-hydroxy-3-methylglutarylcoenzyme A reductase promoters (G¨otz et al 1995, Brion et al 1999). Both linesdeveloped 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

Ann

u. R

ev. N

euro

sci.

2001

.24:

1121

-115

9. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by O

pen

Uni

vers

ity o

n 07

/11/

13. F

or p

erso

nal u

se o

nly.

Page 22: Virginia M-Y Lee,1 Michel Goedert,2 and John Q Trojanowski

P1: FUM

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

1142 LEE ¥ GOEDERT ¥ TROJANOWSKIT

AB

LE

3T

rans

geni

cm

odel

sof

tauo

path

ies

Gen

eP

rom

oter

Tau

Pat

holo

gyP

heno

type

Ref

eren

ce

Tau,

4R/2

NT

hy1

Som

atod

endr

itic

tau

expr

essi

onN

orm

alG

otz

etal

(199

5)

Tau,

3R/0

NH

MG

-CoA

Som

atod

endr

itic

tau

expr

essi

onB

rion

etal

(199

9)re

duct

ase

Tau,

3R/0

NPr

ion

prot

ein

Som

atod

endr

itic

tau

expr

essi

on;t

auA

xono

path

yw

ithIs

hiha

raet

alim

mun

orea

ctiv

esp

hero

ids

inbr

ain

mus

cle

wea

knes

s(1

999,

2001

)an

dsp

inal

cord

;neu

rofib

rilla

ryta

ngle

sin

brai

nat

18m

onth

sor

olde

r

Tau,

4R/2

NT

hy1

Som

atod

endr

itic

tau

expr

essi

on;t

auA

xono

path

yw

ithse

nsor

i-Sp

ittae

lset

alim

mun

orea

ctiv

esp

hero

ids

inbr

ain

mot

ordy

sfun

ctio

n(1

999)

and

spin

alco

rd

Tau,

4R/2

NT

hy1

Som

atod

endr

itic

tau

expr

essi

on;t

auA

xono

path

yw

ithPr

obst

etal

(200

0)im

mun

orea

ctiv

esp

hero

ids

inbr

ain

mus

cle

wea

knes

san

dsp

inal

cord

Tau,

4R/0

N,

Prio

npr

otei

nN

euro

fibri

llary

tang

les

inbr

ain

and

spin

alM

otor

and

beha

vior

alL

ewis

etal

(200

0)P3

01L

cord

;som

atod

endr

itic

tau

expr

essi

onde

ficits

;am

yotr

ophy

Tau,

4R/2

N,

Thy

1N

euro

fibri

llary

tang

les

inbr

ain

and

spin

alG

otz

etal

(200

1)P3

01L

cord

;som

atod

endr

itic

tau

expr

essi

on

Tau,

geno

mic

End

ogen

ous

Axo

nale

xpre

ssio

nN

orm

alD

uff

etal

(200

0)

Apo

E4

Mul

tiple

Phos

phor

ylat

edta

uex

pres

sion

inM

otor

dysf

unct

ion

Tess

eur

etal

(200

0)ne

ocor

tex,

hipp

ocam

pus,

and

amyg

dala

and

amyo

trop

hy

p25

Neu

ron

spec

ific

Phos

phor

ylat

edta

uex

pres

sion

inIn

crea

sed

loco

-A

hlija

nian

enol

ase

cort

ex,a

myg

dala

,and

thal

amus

mot

orac

tivity

etal

(200

0)

Ant

i-N

GF

Cyt

omeg

alov

irus

Phos

phor

ylat

edta

uex

pres

sion

inSp

atia

lmem

ory

and

Cap

soni

IgH

/Igκ

earl

yre

gion

cort

exan

dhi

ppoc

ampu

sw

ithob

ject

reco

gniti

onet

al(2

000)

asso

ciat

edne

uron

loss

impa

irm

ent

Ann

u. R

ev. N

euro

sci.

2001

.24:

1121

-115

9. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by O

pen

Uni

vers

ity o

n 07

/11/

13. F

or p

erso

nal u

se o

nly.

Page 23: Virginia M-Y Lee,1 Michel Goedert,2 and John Q Trojanowski

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 reticulospinalneurons led to the formation of PHF-like tau inclusions with degeneration of asubset of these neurons (Hall et al 1997, 2000). Subsequently, a series of papershas described lines of TG mice expressing high levels of 3R/0N or 4R/2N humantau 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 bodiesand dendrites and large numbers of pathologically enlarged axons containing tau-immunoreactive spheroids. These changes were most prominent in spinal cord butwere also seen in brain. They were accompanied by histological and behavioralsigns of amytrophy. Mice doubly transgenic for 4R2N tau and GSK-3β showedincreased levels of tau phosphorylation and a marked reduction in the number ofspheroids and associated histological and behavioral changes at 3–4 months of age(Spittaels et al 2000). In this system, therefore, hyperphosphorylation of tau corre-lates inversely with pathology. Although the tau pathology most closely resemblesthat observed in the amyotrophic lateral sclerosis/parkinsonism–dementia complex(Matsumoto et al 1990), it differs in several respects from that found in humandiseases. 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 Sand do not bind Gallyas silver. However, a recent report has shown that as thesetau TG mice aged to over 18 months, modest numbers of filamentous tau tangleswith similar properties to those found in AD could be detected in hippocampusand entorhinal cortex (Ishihara et al 2001). Moreover, like NFTs in AD, the tautangles in these aged mice were ubiquitinated, did not contain neurofilaments ortubulin, and were detected by Congo red, Thioflavin S, and Gallyas silver stainingmethods. It is interesting that mice transgenic for the entire humantau gene ex-pressed all six tau isoforms but failed to develop significant pathology (Duff et al2000).

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 tauwith and without the P301L mutation. In contrast to mice expressing WT tau, miceexpressing tau with the P301L mutation exhibited an age- and gene dose-dependentaccumulation of tau tangles in both brain and spinal cord, with associated nerve cellloss and reactive gliosis. Similar results have been reported by G¨otz et al (2001)in TG lines expressing 4R2N tau with the P301L mutation. The tau tangles foundin these mice appeared to comprise only the mutant human tau, which suggeststhat the P301L mutation probably causes neurodegeneration by promoting the ag-gregation of the mutant tau. This is supported by recent biochemical studies usingantibodies specific for P301L tau that demonstrated recovery of mutant but notWT tau from the insoluble fraction isolated from brain tissue of individuals withthe P301Ltaugene mutation (Rizzu et al 2000).

Other approaches to the development of tau pathology in TG mice have madeuse of molecules known to interact with tau. For example, TG mice overexpressing

Ann

u. R

ev. N

euro

sci.

2001

.24:

1121

-115

9. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by O

pen

Uni

vers

ity o

n 07

/11/

13. F

or p

erso

nal u

se o

nly.

Page 24: Virginia M-Y Lee,1 Michel Goedert,2 and John Q Trojanowski

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 architectureand behavioral alterations (Ahlijanian et al 2000). However, there was no biochem-ical evidence of an accumulation of insoluble, hyperphosphorylated tau in thesemice. TG mice expressing apolipoprotein E4, an allelic risk factor for sporadicAD (Corder et al 1993), showed an age-dependent increase in tau phosphorylationthat 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-dependentneurodegenerative pathology, including neuronal loss and hyperphosphorylated,insoluble tau in cortex and hippocampus (Capsoni et al 2000). However, as forthe apolipoprotein E4 TG mice described above, no evidence of fibrillary taupathology was presented. In summary, although several TG mouse models showfeatures of various tau-related disorders, they still fall short of demonstrating theentire 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 distincttopographic and cell type–specific distributions of inclusions. The biochemicaland ultrastructural characteristics of thetau abnormalities, which are frequentlyrelated to the inclusion or exclusion of E10, also reveal a significant phenotypicoverlap. The discovery of multiple mutations in thetau gene that lead to the ab-normal aggregation of tau and cause FTDP-17 demonstrates that tau dysfunctionis sufficient to produce neurodegenerative disease. The mutations lead to specificcellular alterations, including altered expression, function, and biochemistry of tauprotein. The finding that specific polymorphisms and mutations lead to diversephenotypes raises the possibility that the clinical and pathological expression ofthese 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 functionof MTs and interferes with axonal transport. It remains to be established whethera protein kinase/phosphatase imbalance is an early mechanistic step leading to thegeneration of filamentous tau in some tauopathies. Genetic and/or environmentalfactors could initiate a cascade of events that leads to the abnormal phosphorylationof tau through incompletely defined pathways (Figure 4). It also remains to beseen whether the mere presence of tau filaments inside brain cells is sufficient tocause them to die. Similarly, the precise mechanisms by which tau protein assem-bles into filaments in human brain remain to be discovered. Further investigationinto the mechanisms of tau dysfunction, as well as the identification of potentialdisease-modifying factors, will provide additional insight into novel strategies fordisease treatment and prevention. The development of additional animal models

Ann

u. R

ev. N

euro

sci.

2001

.24:

1121

-115

9. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by O

pen

Uni

vers

ity o

n 07

/11/

13. F

or p

erso

nal u

se o

nly.

Page 25: Virginia M-Y Lee,1 Michel Goedert,2 and John Q Trojanowski

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 thisundertaking.

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 offilamentous proteins. Abnormal protein aggregation is observed in a large numberof neurodegenerative disorders (Prusiner 1998, Goedert et al 1998, Trojanowskiet al 1998). Thus, besides tau pathology, AD is characterized by the extracellularaccumulation of Aβ fibrils in the form of amyloid plaques; Lewy body disorderscontain intracytoplasmic filamentous aggregates ofα-synuclein; trinucleotide re-peat disorders have intranuclear inclusions composed of fibrous polyglutamines;and spongiform encephalopathies demonstrate aggregates of proteinase-resistantprion protein. Aggregation of proteins in the brain is a common theme in a diversegroup of disorders, and insight into the pathogenesis of any one of these disordersmay have implications for our understanding of the mechanisms that underlie allthese diseases.

ACKNOWLEDGMENTS

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

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

LITERATURE CITED

Ahlijanian MK, Barrezueta NX, Williams RD,Jakowski A, Kowsz KP, et al. 2000. Hyper-phosphorylated tau and neurofilament andcytoskeletal disruptions in mice overexpress-ing human p25, an activator of cdk5.Proc.Natl. Acad. Sci. USA97(6):2910–15

Andreadis A, Brown WM, Kosik KS. 1992.Structure and novel exons of the human taugene.Biochemistry31(43):10626–33

Arawaka S, Usami M, Sahara N, SchellenbergGD, Lee G, Mori H. 1999. The tau mutation(val337met) disrupts cytoskeletal networksof microtubules.NeuroReport10(5):993–97

Arima K, Kowalska A, Hasegawa M,Mukoyama M, Watanabe R, et al. 2000. Twobrothers with frontotemporal dementia and

parkinsonism with an N279K mutation ofthe tau gene.Neurology54(9):1787–95

Arnold SE, Han LY, Clark CM, Grossman M,Trojanowski JQ. 2000. Quantitative neuro-histological features of frontotemporal de-generation.Neurobiol. Aging21(6):913–919

Arrasate M, Perez M, Armas-Portela R, Avila J.1999. Polymerization of tau peptides into fib-rillar structures. The effect of FTDP-17 mu-tations.FEBS Lett.446(1):199–202

Arrasate M, Perez M, Valpuesta JM, Avila J.1997. Role of glycosaminoglycans in de-termining the helicity of paired helical fil-aments.Am. J. Pathol.151(4):1115–22

Baker M, Litvan I, Houlden H, Adamson J,Dickson D, et al. 1999. Association of an

Ann

u. R

ev. N

euro

sci.

2001

.24:

1121

-115

9. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by O

pen

Uni

vers

ity o

n 07

/11/

13. F

or p

erso

nal u

se o

nly.

Page 26: Virginia M-Y Lee,1 Michel Goedert,2 and John Q Trojanowski

P1: FUM

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

1146 LEE ¥ GOEDERT ¥ TROJANOWSKI

extended haplotype in the tau gene withprogressive supranuclear palsy.Hum. Mol.Genet.8(4):711–15

Barghorn S, Zheng-Fischh¨ofer Q, Ackmann M,Biernat J, von Bergen M, et al. 2000. Struc-ture, microtubule interactions, and paired he-lical filament aggregation by tau mutantsof frontotemporal dementias.Biochemistry39(38):11714–21

Baudier J, Cole RD. 1987. Phosphorylation oftau proteins to a state like that in Alzheimer’sbrain is catalyzed by a calcium/calmodulin-dependent kinase and modulated by phos-pholipids.J. Biol. Chem.262(36):17577–83

Baumann K, Mandelkow EM, Biernat J,Piwnica-Worms H, Mandelkow E. 1993.Abnormal Alzheimer-like phosphorylationof tau-protein by cyclin-dependent kinasescdk2 and cdk5.FEBS Lett.336(3):417–24

Bennett P, Bonifati V, Bonuccelli U, ColosimoC, De Mari M, et al. 1998. Direct genetic ev-idence for involvement of tau in progressivesupranuclear palsy. European Study Groupon Atypical Parkinsonism Consortium.Neu-rology51(4):982–85

Biernat J, Gustke N, Drewes G, MandelkowEM, Mandelkow E. 1993. Phosphorylationof Ser262 strongly reduces binding of tau tomicrotubules: distinction between PHF-likeimmunoreactivity and microtubule binding.Neuron11(1):153–63

Billingsley ML, Kincaid RL. 1997. Regulatedphosphorylation and dephosphorylation oftau protein: effects on microtubule interac-tion, intracellular trafficking and neurode-generation.Biochem. J.323(3):577–91

Binder LI, Frankfurter A, Rebhun LI. 1985. Thedistribution of tau in the mammalian centralnervous system.J. Cell Biol.101(4):1371–78

Bird TD, Nochlin D, Poorkaj P, Cherrier M,Kaye J, et al. 1999. A clinical pathologicalcomparison of three families with frontotem-poral dementia and identical mutations in thetau gene (P301L).Brain 122(4):741–56

Bird TD, Wijsman EM, Nochlin D, LeeheyM, Sumi SM, et al. 1997. Chromosome 17and hereditary dementia: linkage studies in

three nonAlzheimer families and kindredswith late-onset FAD.Neurology48(4):949–54

Bonifati V, Joosse M, Nicholl DJ, VanacoreN, Bennett P, et al. 1999. The tau gene inprogressive supranuclear palsy: exclusion ofmutations in coding exons and exon 10 splicesites, and identification of a new intronic vari-ant of the disease-associated H1 haplotypein Italian cases.Neurosci. Lett.274(1):61–65

Braak E, Braak H, Mandelkow EM. 1994. A se-quence of cytoskeleton changes related to theformation of neurofibrillary tangles and neu-ropil threads.Acta Neuropathol.87(6):554–67

Bramblett GT, Goedert M, Jakes R, Merrick SE,Trojanowski JQ, Lee VM-Y. 1993. Abnormaltau phosphorylation at Ser396 in Alzheimer’sdisease recapitulates development and con-tributes to reduced microtubule binding.Neu-ron 10(6):1089–99

Brion JP, Passareiro H, Nunez J, Flament-Durand J. 1985. Mise en ´evidence im-munologique de la prot´eine tau au niveau deslesions de d´egenerescence neurofibrillaire dela maladie d’Alzheimer.Arch. Biol.95:229–35

Brion JP, Tremp G, Octave JN. 1999. Trans-genic expression of the shortest human tauaffects its compartmentalization and its phos-phorylation as in the pretangle stage ofAlzheimer’s disease.Am. J. Pathol.154(1):255–70

Buee L, Bussi`ere T, Buee-Scherrer V, Dela-courte A, Hof PR. 2000. Tau protein iso-forms, phosphorylation and role in neu-rodegenerative disorders.Brain Res. Rev.33:(1):95–130

Buee-Scherrer V, Hof PR, Bu´ee L, Leveugle B,Vermersch P, et al. 1996. Hyperphosphory-lated tau proteins differentiate corticobasaldegeneration and Pick’s disease.Acta Neu-ropathol.91(4):351–59

Bugiani O, Murrell JR, Giaccone G, HasegawaM, Ghigo G, et al. 1999. Frontotemporal de-mentia and corticobasal degeneration in a

Ann

u. R

ev. N

euro

sci.

2001

.24:

1121

-115

9. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by O

pen

Uni

vers

ity o

n 07

/11/

13. F

or p

erso

nal u

se o

nly.

Page 27: Virginia M-Y Lee,1 Michel Goedert,2 and John Q Trojanowski

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-ropathol. Exp. Neurol.58(6):667–77

Butner KA, Kirschner MW. 1991. Tau proteinbinds to microtubules through a flexible ar-ray of distributed weak sites.J. Cell Biol.115(3):717–30

Capsoni S, Ugolini G, Comparini A, Ruberti F,Berardi N, Cattaneo A. 2000. Alzheimer-likeneurodegeneration in aged antinerve growthfactor transgenic mice.Proc. Natl. Acad. Sci.USA97(12):6826–31

Chambers CB, Lee JM, Troncoso JC, Reich S,Muma NA. 1999. Overexpression of 4R-taumRNA isoforms in progressive supranuclearpalsy but not in Alzheimer’s disease.Ann.Neurol.46(3):325–32

Clark LN, Poorkaj P, Wszolek Z, GeschwindDH, Nasreddine ZS, et al. 1998. Pathogenicimplications of mutations in the tau genein pallido-ponto-nigral degeneration and re-lated neurodegenerative disorders linked tochromosome 17.Proc. Natl. Acad. Sci. USA95(22):13103–7

Cleveland DW, Hwo SY, Kirschner MW. 1977a.Physical and chemical properties of purifiedtau factor and the role of tau in microtubuleassembly.J. Mol. Biol.116(2):227–47

Cleveland DW, Hwo SY, Kirschner MW. 1977b.Purification of tau, a microtubule-associatedprotein that induces assembly of micro-tubules from purified tubulin.J. Mol. Biol.116(2):207–25

Cohen P. 1999. The Croonian Lecture 1998.Identification of a protein kinase cascade ofmajor importance in insulin signal transduc-tion. Philos. Trans. R. Soc. London Ser. B354:485–95

Conrad C, Andreadis A, Trojanowski JQ, Dick-son DW, Kang D, et al. 1997. Genetic ev-idence for the involvement of tau in pro-gressive supranuclear palsy.Ann. Neurol.41(2):277–81

Constantinidis J, Richard J, Tissot R. 1974.Pick’s disease. Histological and clinical cor-relations.Eur. Neurol.11(4):208–17

Corder EH, Saunders AM, Strittmatter WJ,Schmechel DE, Gaskell PC, et al. 1993. Gene

dose of apolipoprotein E type 4 allele and therisk of Alzheimer’s disease in late onset fam-ilies. Science261(5123):921–23

Couchie D, Mavilia C, Georgieff IS, Liem RK,Shelanski ML, Nunez J. 1992. Primary struc-ture of high molecular weight tau present inthe peripheral nervous system.Proc. Natl.Acad. Sci. USA89(10):4378–81

Crowther RA. 1991. Straight and paired helicalfilaments in Alzheimer disease have a com-mon structural unit.Proc. Natl. Acad. Sci.USA88(6):2288–92

Crowther RA, Goedert M. 2000. Abnormaltau-containing filaments in neurodegenera-tive diseases.J. Struct. Biol.130(2–3):271–79

Crowther RA, Olesen OF, Jakes R, GoedertM. 1992. The microtubule binding repeatsof tau protein assemble into filaments likethose found in Alzheimer’s disease.FEBSLett.309(2):199–202

Crowther RA, Wischik CM. 1985. Image re-construction of the Alzheimer paired helicalfilament.EMBO J.4(13B):3661–65

Dayanandan R, van Slegtenhorst M, Mack TG,Ko L, Yen SH, et al. 1999. Mutations in taureduce its microtubule binding properties inintact cells and affect its phosphorylation.FEBS Lett.446(2–3):228–32

Delacourte A, Robitaille Y, Sergeant N, Bu´ee L,Hof PR, et al. 1996. Specific pathological Tauprotein variants characterize Pick’s disease.J. Neuropathol. Exp. Neurol.55(2):159–68

Delacourte A, Sergeant N, Wattez A, GauvreauD, Robitaille Y. 1998. Vulnerable neuronalsubsets in Alzheimer’s and Pick’s diseaseare distinguished by their tau isoform dis-tribution and phosphorylation.Ann. Neurol.43(2):193–204

Delisle MB, Murrell JR, Richardson R, Tro-fatter JA, Rascol O, et al. 1999. A muta-tion at codon 279 (N279K) in exon 10 ofthe Tau gene causes a tauopathy with demen-tia and supranuclear palsy.Acta Neuropathol.98(1):62–77

DeTure M, Ko LW, Yen S, Nacharaju P, Eas-son C, et al. 2000. Missense tau mutations

Ann

u. R

ev. N

euro

sci.

2001

.24:

1121

-115

9. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by O

pen

Uni

vers

ity o

n 07

/11/

13. F

or p

erso

nal u

se o

nly.

Page 28: Virginia M-Y Lee,1 Michel Goedert,2 and John Q Trojanowski

P1: FUM

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

1148 LEE ¥ GOEDERT ¥ TROJANOWSKI

identified in FTDP-17 have a small effecton tau-microtubule interactions.Brain Res.853(1):5–14

Dickson DW. 1998. Pick’s disease: a modernapproach.Brain Pathol.8(2):339–54

Di Maria E, Tabaton M, Vigo T, AbbruzzeseG, Bellone E, et al. 2000. Corticobasal de-generation shares a common genetic back-ground with progressive supranuclear palsy.Ann. Neurol.47(3):374–77

Drechsel DN, Hyman AA, Cobb MH,Kirschner MW. 1992. Modulation of thedynamic instability of tubulin assembly bythe microtubule-associated protein tau.Mol.Biol. Cell 3(10):1141–54

Drewes G, Ebneth A, Preuss U, MandelkowEM, Mandelkow E. 1997. MARK, a novelfamily of protein kinases that phosphory-late microtubule-associated proteins and trig-ger microtubule disruption.Cell 89(2):297–308

Drewes G, Lichtenberg-Kraag B, D¨oring F,Mandelkow EM, Biernat J, et al. 1992. Mi-togen activated protein (MAP) kinase trans-forms tau protein into an Alzheimer-likestate.EMBO J.11(6):2131–38

Drewes G, Mandelkow EM, Baumann K,Goris J, Merlevede W, Mandelkow E.1993. Dephosphorylation of tau protein andAlzheimer paired helical filaments by cal-cineurin and phosphatase-2A.FEBS Lett.336(3):425–32

D’Souza I, Poorkaj P, Hong M, Nochlin D, LeeVM-Y, et al. 1999. Missense and silent taugene mutations cause frontotemporal demen-tia with parkinsonism-chromosome 17 type,by affecting multiple alternative RNA splic-ing regulatory elements.Proc. Natl. Acad.Sci. USA96(10):5598–603

D’Souza I, Schellenberg D. 2000. Determinantsof 4 repeat tau expression: coordination be-tween enhancing and inhibitory splicing se-quences for exon 10 inclusion.J. Biol. Chem.275(23):17700–9

Duff K, Knight H, Refolo LM, Sanders S, Yu X,et al. 2000. Characterization of pathology intransgenic mice over-expressing human ge-

nomic and cDNA tau transgenes.Neurobiol.Dis. 7(2):87–98

Dumanchin C, Camuzat A, Campion D, Verpil-lat P, Hannequin D, et al. 1998. Segregationof a missense mutation in the microtubule-associated protein tau gene with familialfrontotemporal dementia and parkinsonism.Hum. Mol. Genet.7(11):1825–29

Ezquerra M, Pastor P, Valldeoriola F, Molin-uevo JL, Blesa R, et al. 1999. Identification ofa novel polymorphism in the promoter regionof the tau gene highly associated to progres-sive supranuclear palsy in humans.Neurosci.Lett.275(3):183–86

Feany MB, Dickson DW. 1995. Widespreadcytoskeletal pathology characterizes cor-ticobasal degeneration.Am. J. Pathol.146(6):1388–96

Feany MB, Ksiezak-Reding H, Liu WK, Vin-cent I, Yen SH, Dickson DW. 1995. Epi-tope expression and hyperphosphorylation oftau protein in corticobasal degeneration: dif-ferentiation from progressive supranuclearpalsy.Acta Neuropathol.90(1):37–43

Feany MB, Mattiace LA, Dickson DW.1996. Neuropathologic overlap of progres-sive supranuclear palsy, Pick’s disease andcorticobasal degeneration.J. Neuropathol.Exp. Neurol.55(1):53–67

Flament S, Delacourte A, Verny M, Hauw JJ,Javoy-Agid F. 1991. Abnormal tau proteinsin progressive supranuclear palsy. Similari-ties and differences with the neurofibrillarydegeneration of the Alzheimer type.ActaNeuropathol.81(6):591–96

Foster NL, Wilhelmsen K, Sima AA, Jones MZ,D’Amato CJ, et al. 1997. Frontotemporal de-mentia and parkinsonism linked to chromo-some 17: a consensus conference.Ann. Neu-rol. 41(6):706–15

Frappier T, Liang NS, Brown K, Leung CL,Lynch T, et al. 1999. Abnormal microtubulepacking in processes of SF9 cells express-ing the FTDP-17 V337M tau mutation.FEBSLett.455(3):262–66

Friedhoff P, von Bergen M, Mandelkow EM,Davies P, Mandelkow E. 1998. A nucleated

Ann

u. R

ev. N

euro

sci.

2001

.24:

1121

-115

9. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by O

pen

Uni

vers

ity o

n 07

/11/

13. F

or p

erso

nal u

se o

nly.

Page 29: Virginia M-Y Lee,1 Michel Goedert,2 and John Q Trojanowski

P1: FUM

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

NEW INSIGHTS INTO TAUOPATHIES 1149

assembly mechanism of Alzheimer pairedhelical filaments.Proc. Natl. Acad. Sci. USA95(26):15712–17

Gamblin TC, King ME, Dawson H, Vitek MP,Kuret J, et al. 2000. In vitro polymeriza-tion of tau protein monitored by laser lightscattering: method and application to thestudy of FTDP-17 mutants.Biochemistry39(20):6136–44

Games D, Adams D, Alessandrini R, BarbourR, Berthelette P, et al. 1995. Alzheimer-typeneuropathology in transgenic mice overex-pressing V717F beta-amyloid precursor pro-tein.Nature373(6514):523–27

Gao QS, Memmott J, Lafyatis R, Stamm S,Sreaton G, Andreadis A. 2000. Complexregulation of tau exon 10, whose missplic-ing causes frontotemporal dementia.J. Neu-rochem.74(2):490–500

Georgieff IS, Liem RK, Mellado W, Nunez J,Shelanski ML. 1991. High molecular weighttau: preferential localization in the periph-eral nervous system.J. Cell Sci.100(1):55–60

Ginsberg SD, Galvin JE, Chiu TS, Lee VM-Y, Masliah E, Trojanowski JQ. 1998. RNAsequestration to pathological lesions of neu-rodegenerative diseases.Acta Neuropathol.96(5):487–94

Goedert M, Cohen ES, Jakes R, Cohen P. 1992a.p42 MAP kinase phosphorylation sites inmicrotubule-associated protein tau are de-phosphorylated by protein phosphatase 2A1.Implications for Alzheimer’s disease.FEBSLett.312(1):95–99

Goedert M, Hasegawa M, Jakes R, LawlerS, Cuenda A, Cohen P. 1997. Phosphoryla-tion of microtubule-associated protein tau bystress-activated protein kinases.FEBS Lett.409(1):57–62

Goedert M, Jakes R. 1990. Expression of sep-arate isoforms of human tau protein: corre-lation with the tau pattern in brain and ef-fects on tubulin polymerization.EMBO J.9(13):4225–30

Goedert M, Jakes R, Crowther RA. 1999a. Ef-fects of frontotemporal dementia FTDP-17

mutations on heparin-induced assembly oftau filaments.FEBS Lett.450(3):306–11

Goedert M, Jakes R, Crowther RA, Six J, L¨ubkeU, et al. 1993. The abnormal phosphoryla-tion of tau protein at Ser202 in Alzheimerdisease recapitulates phosphorylation duringdevelopment.Proc. Natl. Acad. Sci. USA90(11):5066–70

Goedert M, Jakes R, Qi Z, Wang JH, Cohen P.1995a. Protein phosphatase 2A is the majorenzyme in brain that dephosphorylates tauprotein phosphorylated by proline-directedprotein kinases or cyclic AMP-dependentprotein kinase.J. Neurochem.65(6):2804–7

Goedert M, Jakes R, Spillantini MG, HasegawaM, Smith MJ, Crowther RA. 1996. As-sembly of microtubule-associated proteintau into Alzheimer-like filaments inducedby sulphated glycosaminoglycans.Nature383(6600):550–53

Goedert M, Satumtira S, Jakes R, Smith MJ,Kamibayashi C, et al. 2000. Reduced bindingof protein phosphatase 2A to tau protein withfrontotemporal dementia and parkinsonismlinked to chromosome 17 mutations.J. Neu-rochem.75(5):2155–62

Goedert M, Spillantini MG, Cairns NJ,Crowther RA. 1992b. Tau proteins ofAlzheimer paired helical filaments: abnor-mal phosphorylation of all six brain isoforms.Neuron8(1):159–68

Goedert M, Spillantini MG, Crowther RA.1992c. Cloning of a big tau microtubule-associated protein characteristic of the pe-ripheral nervous system.Proc. Natl. Acad.Sci. USA89(5):1983–87

Goedert M, Spillantini MG, Crowther RA,Chen SG, Parchi P, et al. 1999b. Tau genemutation in familial progressive subcorticalgliosis.Nat. Med.5(4):454–57

Goedert M, Spillantini MG, Davies SW. 1998.Filamentous nerve cell inclusions in neurode-generative diseases.Curr. Opin. Neurobiol.8(5):619–32

Goedert M, Spillantini MG, Jakes R, CrowtherRA, Vanmechelen E, et al. 1995b. Molecular

Ann

u. R

ev. N

euro

sci.

2001

.24:

1121

-115

9. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by O

pen

Uni

vers

ity o

n 07

/11/

13. F

or p

erso

nal u

se o

nly.

Page 30: Virginia M-Y Lee,1 Michel Goedert,2 and John Q Trojanowski

P1: FUM

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

1150 LEE ¥ GOEDERT ¥ TROJANOWSKI

dissection of the paired helical filament.Neu-robiol. Aging16(3):325–34

Goedert M, Spillantini MG, Jakes R, Ruther-ford D, Crowther RA. 1989a. Multipleisoforms of human microtubule-associatedprotein tau: sequences and localization inneurofibrillary tangles of Alzheimer’s dis-ease.Neuron3(4):519–26

Goedert M, Spillantini MG, Potier MC, Ul-rich J, Crowther RA. 1989b. Cloning andsequencing of the cDNA encoding an iso-form of microtubule-associated protein taucontaining four tandem repeats: differentialexpression of tau protein mRNAs in humanbrain.EMBO J.8(2):393–99

Goedert M, Wischik CM, Crowther RA, WalkerJE, Klug A. 1988. Cloning and sequenc-ing of the cDNA encoding a core proteinof the paired helical filament of Alzheimerdisease: identification as the microtubule-associated protein tau.Proc. Natl. Acad. Sci.USA85(11):4051–55

Goode BL, Denis PE, Panda D, Radeke MJ,Miller HP, et al. 1997. Functional interactionsbetween the proline-rich and repeat regionsof tau enhance microtubule binding and as-sembly.Mol. Biol. Cell8(2):353–65

Goode BL, Feinstein SC. 1994. Identificationof a novel microtubule binding and assem-bly domain in the developmentally regu-lated interrepeat region of tau.J. Cell Biol.124(5):769–82

Gotz J, Chen F, Barmettler R, Nitsch RM.2001. Tau filament formation in transgenicmice expressing P301L tau.J. Biol. Chem.276(1):529–34

Gotz J, Probst A, Spillantini MG, Schafer T,Jakes R, et al. 1995. Somatodendritic lo-calization and hyperphosphorylation of tauprotein in transgenic mice expressing thelongest human brain tau isoform.EMBO J.14(7):1304–13

Greenberg SG, Davies P. 1990. A preparation ofAlzheimer paired helical filaments that dis-plays distinct tau proteins by polyacrylamidegel electrophoresis.Proc. Natl. Acad. Sci.USA87(15):5827–31

Greenberg SG, Davies P, Schein JD, Binder LI.1992. Hydrofluoric acid-treated tau PHF pro-teins display the same biochemical propertiesas normal tau.J. Biol. Chem.267(1):564–69

Grover A, Houlden H, Baker M, Adamson J,Lewis J, et al. 1999. 5′ splice site mutationsin tau associated with the inherited demen-tia FTDP-17 affect a stem-loop structure thatregulates alternative splicing of exon 10.J.Biol. Chem.274(21):15134–43

Grundke-Iqbal I, Iqbal K, Tung YC, QuinlanM, Wisniewski HM, Binder LI. 1986. Ab-normal phosphorylation of the microtubule-associated protein tau (tau) in Alzheimer cy-toskeletal pathology.Proc. Natl. Acad. Sci.USA83(13):4913–17

Gustke N, Trinczek B, Biernat J, MandelkowEM, Mandelkow E. 1994. Domains of tauprotein and interactions with microtubules.Biochemistry33(32):9511–22

Hall GF, Chu B, Lee G, Yao J. 2000. Humantau filaments induce microtubule and synapseloss in an in vivo model of neurofibrillary de-generative disease.J. Cell Sci.113(8):1373–87

Hall GF, Yao J, Lee G. 1997. Human tau be-comes phosphorylated and forms filamen-tous deposits when overexpressed in lampreycentral neurons in situ.Proc. Natl. Acad. Sci.USA94(9):4733–38

Hanger DP, Hughes K, Woodgett JR, BrionJP, Anderton BH. 1992. Glycogen syn-thase kinase-3 induces Alzheimer’s disease-like phosphorylation of tau: generation ofpaired helical filament epitopes and neuronallocalisation of the kinase.Neurosci. Lett.147(1):58–62

Harada A, Oguchi K, Okabe S, Kuno J, TeradaS, et al. 1994. Altered microtubule organiza-tion in small-calibre axons of mice lackingtau protein.Nature369(6480):488–91

Hardy J, Allsop D. 1991. Amyloid deposi-tion as the central event in the aetiology ofAlzheimer’s disease.Trends Pharmacol. Sci.12(10):383–88

Hasegawa M, Crowther RA, Jakes R, Goed-ert M. 1997. Alzheimer-like changes in

Ann

u. R

ev. N

euro

sci.

2001

.24:

1121

-115

9. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by O

pen

Uni

vers

ity o

n 07

/11/

13. F

or p

erso

nal u

se o

nly.

Page 31: Virginia M-Y Lee,1 Michel Goedert,2 and John Q Trojanowski

P1: FUM

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

NEW INSIGHTS INTO TAUOPATHIES 1151

microtubule-associated protein tau inducedby sulfated glycosaminoglycans. Inhibitionof microtubule binding, stimulation of phos-phorylation, and filament assembly dependon the degree of sulfation.J. Biol. Chem.272(52):33118–24

Hasegawa M, Jakes R, Crowther RA, LeeVM-Y, Ihara Y, Goedert M. 1996. Char-acterization of mAb AP422, a novelphosphorylation-dependent monoclonal an-tibody against tau protein.FEBS Lett.384(1):25–30

Hasegawa M, Smith MJ, Goedert M. 1998. Tauproteins with FTDP-17 mutations have a re-duced ability to promote microtubule assem-bly. FEBS Lett.437(3):207–10

Hasegawa M, Smith MJ, Iijima M, TabiraT, Goedert M. 1999. FTDP-17 mutationsN279K and S305N in tau produce increasedsplicing of exon 10.FEBS Lett.443(2):93–96

Hauw JJ, Daniel SE, Dickson D, Horou-pian DS, Jellinger K, et al. 1994. Pre-liminary NINDS neuropathologic criteriafor Steele-Richardson-Olszewski syndrome(progressive supranuclear palsy).Neurology44(11):2015–19

Hauw JJ, Verny M, Delaere P, Cervera P, HeY, Duyckaerts C. 1990. Constant neurofib-rillary changes in the neocortex in progres-sive supranuclear palsy. Basic differenceswith Alzheimer’s disease and aging.Neu-rosci. Lett.119(2):182–86

Heutink P, Stevens M, Rizzu P, Bakker E,Kros JM, et al. 1997. Hereditary frontotem-poral dementia is linked to chromosome17q21-q22: a genetic and clinicopatholog-ical study of three Dutch families.Ann. Neu-rol. 41(2):150–59

Hiesberger T, Trommsdorff M, Howell BW,Goffinet A, Mumby MC, et al. 1999. Di-rect binding of Reelin to VLDL receptor andApoE receptor 2 induces tyrosine phosphory-lation of disabled-1 and modulates tau phos-phorylation.Neuron24(2):481–89

Higgins JJ, Litvan I, Pho LT, Li W, Nee LE.1998. Progressive supranuclear gaze palsyis in linkage disequilibrium with the tau

and not the alpha-synuclein gene.Neurology50(1):270–73

Himmler A, Drechsel D, Kirschner MW, MartinDW Jr. 1989. Tau consists of a set of proteinswith repeated C-terminal microtubule-binding domains and variable N-terminaldomains. Mol. Cell Biol. 9(4):1381–88

Hoenicka J, Perez M, Perez-Tur J, Barabash A,Godoy M, et al. 1999. The tau gene A0 alleleand progressive supranuclear palsy.Neurol-ogy53(6):1219–25

Hoffmann R, Lee VM-Y, Leight S, Varga I,Otvos L Jr. 1997. Unique Alzheimer’s dis-ease paired helical filament specific epitopesinvolve double phosphorylation at specificsites.Biochemistry36(26):8114–24

Hong M, Chen DC, Klein PS, Lee VM-Y. 1997.Lithium reduces tau phosphorylation by inhi-bition of glycogen synthase kinase-3.J. Biol.Chem.272(40):25326–32

Hong M, Lee VM-Y. 1997. Insulin and insulin-like growth factor-1 regulate tau phospho-rylation in cultured human neurons.J. Biol.Chem.272(31):19547–53

Hong M, Trojanowski JQ, Lee VM-Y. 2000.Tau-based neurofibrillary lesions. InNeu-rodegenerative Dementias, ed. CM Clark, JQTrojanowski, pp. 161–75. New York: Mc-Graw Hill

Hong M, Zhukareva V, Vogelsberg-Ragaglia V,Wszolek Z, Reed L, et al. 1998. Mutation-specific functional impairments in distincttau isoforms of hereditary FTDP-17.Science282(5395):1914–17

Houlden H, Baker M, Adamson J, Grover A,Waring S, et al. 1999. Frequency of taumutations in three series of non-Alzheimer’sdegenerative dementia.Ann. Neurol.46(2):243–48

Hsiao K, Chapman P, Nilsen S, Eckman C,Harigaya Y, et al. 1996. Correlative memorydeficits, Aβ elevation, and amyloid plaquesin transgenic mice.Science274(5284):99–102

Hulette CM, Pericak-Vance MA, Roses AD,Schmechel DE, Yamaoka LH, et al. 1999.

Ann

u. R

ev. N

euro

sci.

2001

.24:

1121

-115

9. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by O

pen

Uni

vers

ity o

n 07

/11/

13. F

or p

erso

nal u

se o

nly.

Page 32: Virginia M-Y Lee,1 Michel Goedert,2 and John Q Trojanowski

P1: FUM

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

1152 LEE ¥ GOEDERT ¥ TROJANOWSKI

Neuropathological features of frontotempo-ral dementia and parkinsonism linked tochromosome 17q21–22 (FTDP-17): DukeFamily 1684.J. Neuropathol. Exp. Neurol.58(8):859–66

Hutton M, Lendon CL, Rizzu P, Baker M,Froelich S, et al. 1998. Association of mis-sense and 5′-splice-site mutations in tauwith the inherited dementia FTDP-17.Na-ture393(6686):702–5

Iijima M, Tabira T, Poorkaj P, Schellenberg GD,Trojanowski JQ, et al. 1999. A distinct fa-milial presenile dementia with a novel mis-sense mutation in the tau gene.NeuroReport10(3):497–501

Ikegami S, Harada A, Hirokawa N. 2000. Mus-cle weakness, hyperactivity, and impairmentin fear conditioning in tau-deficient mice.Neurosci. Lett.279(3):129–32

Ishiguro K, Kobayashi S, Omori A, TakamatsuM, Yonekura S, et al. 1994. Identification ofthe 23 kDa subunit of tau protein kinase II asa putative activator of cdk5 in bovine brain.FEBS Lett.342(2):203–8

Ishihara T, Hong M, Zhang B, Nakagawa Y, LeeMK, et al. 1999. Age-dependent emergenceand progression of a tauopathy in transgenicmice overexpressing the shortest human tauisoform.Neuron24(3):751–62

Ishihara T, Zhang B, Higuchi M, Yashiyama Y,Trojanowski JQ, Lee VM-Y. 2001. Age de-pendent induction of congophilic neurofib-rillary inclusions in tau transgenic mice.Am.J. Pathol.158(2):555–62

Iwatsubo T, Hasegawa M, Ihara Y. 1994. Neu-ronal and glial tau-positive inclusions indiverse neurologic diseases share commonphosphorylation characteristics.Acta Neu-ropathol.88(2):129–36

Jiang Z, Cote J, Kwon JM, Goate AM, Wu JY.2000. Aberrant splicing of tau pre-mRNAcaused by intronic mutations associated withthe inherited dementia frontotemporal de-mentia with parkinsonism linked to chromo-some 17.Mol. Cell Biol.20(11):4036–48

Kampers T, Friedhoff P, Biernat J, MandelkowEM, Mandelkow E. 1996. RNA stimulates

aggregation of microtubule-associated pro-tein tau into Alzheimer-like paired helical fil-aments.FEBS Lett.399(3):344–49

Kanemaru K, Takio K, Miura R, Titani K, IharaY. 1992. Fetal-type phosphorylation of thetau in paired helical filaments.J. Neurochem.58(5):1667–75

Kidd M. 1963. Paired helical filaments in elec-tron microscopy of Alzheimer’s disease.Na-ture197:192–94

Kobayashi S, Ishiguro K, Omori A, TakamatsuM, Arioka M, et al. 1993. A cdc2-relatedkinase PSSALRE/cdk5 is homologous withthe 30 kDa subunit of tau protein kinase II,a proline-directed protein kinase associatedwith microtubule.FEBS Lett.335(2):171–75

Kodama K, Okada S, Iseki E, Kowalska A,Tabira T, et al. 2000. Familial frontotempo-ral dementia with a P301L tau mutation inJapan.J. Neurol. Sci.176(1):57–64

Komori T. 1999. Tau-positive glial inclusionsin progressive supranuclear palsy, corti-cobasal degeneration and Pick’s disease.Brain Pathol.9(4):663–79

Komori T, Arai N, Oda M, Nakayama H, MoriH, et al. 1998. Astrocytic plaques and tufts ofabnormal fibers do not coexist in corticobasaldegeneration and progressive supranuclearpalsy.Acta Neuropathol.96(4):401–8

Kondo J, Honda T, Mori H, Hamada Y, MiuraR, et al. 1988. The carboxyl third of tauis tightly bound to paired helical filaments.Neuron1(9):827–34

Kosik KS, Orecchio LD, Binder L, TrojanowskiJQ, Lee VM-Y, Lee G. 1988. Epitopes thatspan the tau molecule are shared with pairedhelical filaments.Neuron1(9):817–25

Ksiezak-Reding H, Morgan K, Mattiace LA,Davies P, Liu WK, et al. 1994. Ultrastructureand biochemical composition of paired he-lical filaments in corticobasal degeneration.Am. J. Pathol.145(6):1496–508

Kusakawa G, Saito T, Onuki R, Ishiguro K,Kishimoto T, Hisanaga S. 2000. Calpain-dependent proteolytic cleavage of the p35cyclin-dependent kinase 5 activator to p25.J. Biol. Chem.275(22):17166–72

Ann

u. R

ev. N

euro

sci.

2001

.24:

1121

-115

9. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by O

pen

Uni

vers

ity o

n 07

/11/

13. F

or p

erso

nal u

se o

nly.

Page 33: Virginia M-Y Lee,1 Michel Goedert,2 and John Q Trojanowski

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-tubule binding domain of tau protein.Neuron2(6):1615–24

Lee MS, Kwon YT, Li M, Peng J, FriedlanderRM, Tsai LH. 2000. Neurotoxicity inducescleavage of p35 to p25 by calpain.Nature405(6784):360–64

Lee VM-Y, Balin BJ, Otvos L Jr, TrojanowskiJQ. 1991. A68: a major subunit of paired he-lical filaments and derivatized forms of nor-mal tau.Science251(4994):675–78

Lendon CL, Lynch T, Norton J, McKeel DW Jr,Busfield F, et al. 1998. Hereditary dyspha-sic disinhibition dementia: a frontotempo-ral dementia linked to 17q21–22.Neurology50(6):1546–55

Leost M, Schultz C, Link A, Wu YZ, BiernatJ, et al. 2000. Paullones are potent inhibitorsof glycogen synthase kinase-3β and cyclin-dependent kinase 5/p25.Eur. J. Biochem.267(19):5983–94

Lew J, Huang QQ, Qi Z, Winkfein RJ, Ae-bersold R, et al. 1994. A brain-specific ac-tivator of cyclin-dependent kinase 5.Nature371(6496):423–26

Lew J, Wang JH. 1994. Neuronal cdc2-like ki-nase.Trends Biochem. Sci.20:33–37

Lewis J, McGowan E, Rockwood J, Melrose H,Nacharaju P, et al. 2000. Neurofibrillary tan-gles, amyotrophy and progressive motor dis-turbance in mice expressing mutant (P301L)tau protein.Nat. Genet.25(4):402–5

Liao H, Li Y, Brautigan DL, Gundersen GG.1998. Protein phosphatase 1 is targeted tomicrotubules by the microtubule-associatedprotein tau.J. Biol. Chem.273(34):21901–8

Lieberman AP, Trojanowski JQ, Lee VM-Y,Balin BJ, Ding XS, et al. 1998. Cognitive,neuroimaging, and pathological studies ina patient with Pick’s disease.Ann. Neurol.43(2):259–65

Lippa CF, Zhukareva V, Kawarai T, UryuH, Shafiq M, et al. 2000. Frontotemporaldementia with novel tau pathology and aGlu342Valtau gene mutation.Ann. Neurol.48(6):850–58

Litersky JM, Johnson GV. 1992. Phosphoryla-

tion by cAMP-dependent protein kinase in-hibits the degradation of tau by calpain.J.Biol. Chem.267(3):1563–68

Litvan I, Agid Y, Calne D, Campbell G, DuboisB, et al. 1996. Clinical research criteriafor the diagnosis of progressive supranu-clear palsy (Steele-Richardson-Olszewskisyndrome): report of the NINDS-SPSP in-ternational workshop.Neurology47(1):1–9

LoPresti P, Szuchet S, Papasozomenos SC,Zinkowski RP, Binder LI. 1995. Functionalimplications for the microtubule-associatedprotein tau: localization in oligodendrocytes.Proc. Natl. Acad. Sci. USA92(22):10369–73

Lovestone S, Davis DR, Webster MT, KaechS, Brion JP, et al. 1999. Lithium reduces tauphosphorylation: effects in living cells and inneurons at therapeutic concentrations.Biol.Psychiatry45(8):995–1003

Lovestone S, Hartley CL, Pearce J, AndertonBH. 1996. Phosphorylation of tau by glyco-gen synthase kinase-3 beta in intact mam-malian cells: the effects on the organizationand stability of microtubules.Neuroscience73(4):1145–57

Lu PJ, Wulf G, Zhou XZ, Davies P, Lu KP. 1999.The prolyl isomerase Pin1 restores the func-tion of Alzheimer-associated phosphorylatedtau protein.Nature399(6738):784–88

Lund and Manchester Groups. 1994. Clinicaland neuropathological criteria for frontotem-poral dementia. The Lund and ManchesterGroups. J. Neurol. Neurosurg. Psychiatry57(4):416–18

Mailliot C, Sergeant N, Bussi`ere T, Caillet-Boudin ML, Delacourte A, Bu´ee L. 1998.Phosphorylation of specific sets of tau iso-forms reflects different neurofibrillary degen-eration processes.FEBS Lett.433(3):201–4

Mandelkow EM, Drewes G, Biernat J, GustkeN, Van Lint J, et al. 1992. Glycogen syn-thase kinase-3 and the Alzheimer-like stateof microtubule-associated protein tau.FEBSLett.314(3):315–21

Mann DM. 1998. Dementia of frontal typeand dementias with subcortical gliosis.BrainPathol.8(2):325–38

Ann

u. R

ev. N

euro

sci.

2001

.24:

1121

-115

9. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by O

pen

Uni

vers

ity o

n 07

/11/

13. F

or p

erso

nal u

se o

nly.

Page 34: Virginia M-Y Lee,1 Michel Goedert,2 and John Q Trojanowski

P1: FUM

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

1154 LEE ¥ GOEDERT ¥ TROJANOWSKI

Matsumoto S, Hirano A, Goto S. 1990.Spinal cord neurofibrillary tangles ofGuamanian amyotrophic lateral sclerosisand parkinsonism-dementia complex: animmunohistochemical study. Neurology40(6):975–79

Matsumura N, Yamazaki T, Ihara Y. 1999.Stable expression in Chinese hamster ovarycells of mutated tau genes causing frontotem-poral dementia and parkinsonism linked tochromosome 17 (FTDP-17).Am. J. Pathol.154(6):1649–56

Matsuo ES, Shin RW, Billingsley ML, Van de-Voorde A, O’Connor M, et al. 1994. Biopsy-derived adult human brain tau is phosphory-lated at many of the same sites as Alzheimer’sdisease paired helical filament tau.Neuron13(4):989–1002

Mawal-Dewan M, Henley J, Van de Voorde A,Trojanowski JQ, Lee VM-Y. 1994. The phos-phorylation state of tau in the developing ratbrain is regulated by phosphoprotein phos-phatases.J. Biol. Chem.269(49):30981–87

Merrick SE, Trojanowski JQ, Lee VM-Y. 1997.Selective destruction of stable microtubulesand axons by inhibitors of protein ser-ine/threonine phosphatases in cultured hu-man neurons.J. Neurosci.17(15):5726–37

Mirra SS, Murrell JR, Gearing M, SpillantiniMG, Goedert M, et al. 1999. Tau pathologyin a family with dementia and a P301L mu-tation in tau.J. Neuropathol. Exp. Neurol.58(4):335–45

Mori H, Nishimura M, Namba Y, Oda M. 1994.Corticobasal degeneration: a disease withwidespread appearance of abnormal tau andneurofibrillary tangles, and its relation toprogressive supranuclear palsy.Acta Neu-ropathol.88(2):113–21

Morishima-Kawashima M, Hasegawa M, TakioK, Suzuki M, Yoshida H, et al. 1995. Hy-perphosphorylation of tau in PHF.Neurobiol.Aging16(3):365–71

Morris HR, Janssen JC, Bandmann O, DanielSE, Rossor MN, et al. 1999a. The tau geneA0 polymorphism in progressive supranu-clear palsy and related neurodegenerative

diseases.J. Neurol. Neurosurg. Psychiatry66(5):665–67

Morris HR, Perez-Tur J, Janssen JC, BrownJ, Lees AJ, et al. 1999b. Mutation in thetau exon 10 splice site region in famil-ial frontotemporal dementia.Ann. Neurol.45(2):270–71

Munoz-Garcia D, Ludwin SK. 1984. Classicand generalized variants of Pick’s disease: aclinicopathological, ultrastructural, and im-munocytochemical comparative study.Ann.Neurol.16(4):467–80

Munoz-Montano JR, Moreno FJ, Avila J, Diaz-Nido J. 1997. Lithium inhibits Alzheimer’sdisease-like tau protein phosphorylationin neurons. FEBS Lett. 411(2–3):183–88

Murayama S, Mori H, Ihara Y, Tomonaga M.1990. Immunocytochemical and ultrastruc-tural studies of Pick’s disease.Ann. Neurol.27(4):394–405

Murrell JR, Koller D, Foroud T, GoedertM, Spillantini MG, et al. 1997. Familialmultiple-system tauopathy with presenile de-mentia is localized to chromosome 17.Am.J. Hum. Genet.61(5):1131–38

Murrell JR, Spillantini MG, Zolo P, GuazzelliM, Smith MJ, et al. 1999. Tau gene muta-tion G389R causes a tauopathy with abun-dant Pick body-like inclusions and ax-onal deposits.J. Neuropathol. Exp. Neurol.58(12):1207–26

Nacharaju P, Lewis J, Easson C, Yen S, HackettJ, et al. 1999. Accelerated filament formationfrom tau protein with specific FTDP-17 mis-sense mutations.FEBS Lett.447(2–3):195–99

Nasreddine ZS, Loginov M, Clark LN,Lamarche J, Miller BL, et al. 1999. Fromgenotype to phenotype: a clinical patho-logical, and biochemical investigation offrontotemporal dementia and parkinsonism(FTDP-17) caused by the P301L tau muta-tion. Ann. Neurol.45(6):704–15

Nath R, Davis M, Probert AW, Kupina NC, RenX, et al. 2000. Processing of cdk5 activatorp35 to its truncated form (p25) by calpain

Ann

u. R

ev. N

euro

sci.

2001

.24:

1121

-115

9. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by O

pen

Uni

vers

ity o

n 07

/11/

13. F

or p

erso

nal u

se o

nly.

Page 35: Virginia M-Y Lee,1 Michel Goedert,2 and John Q Trojanowski

P1: FUM

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

NEW INSIGHTS INTO TAUOPATHIES 1155

in acutely injured neuronal cells.Biochem.Biophys. Res. Commun.274(1):16–21

Neve RL, Harris P, Kosik KS, Kurnit DM,Donlon TA. 1986. Identification of cDNAclones for the human microtubule-associatedprotein tau and chromosomal localization ofthe genes for tau and microtubule-associatedprotein 2.Brain Res.387(3):271–80

Patrick GN, Zukerberg L, Nikolic M, de laMonte S, Dikkes P, Tsai LH. 1999. Conver-sion of p35 to p25 deregulates Cdk5 activ-ity and promotes neurodegeneration.Nature402(6762):615–22

Perez M, Lim F, Arrasate M, Avila J. 2000. TheFTDP-17-linked mutation R406W abolishesthe interaction of phosphorylated tau withmicrotubules.J. Neurochem.74(6):2583–89

Perez M, Valpuesta JM, Medina M, Montejo G,Avila J. 1996. Polymerization of tau into fil-aments in the presence of heparin: the mini-mal sequence required for tau-tau interaction.J. Neurochem.67(3):1183–90

Pickering-Brown S, Baker M, Yen SH, Liu WK,Hasegawa M, et al. 2000. Pick’s disease is as-sociated with mutations in thetaugene.Ann.Neurol.48(5):806–8

Pollock NJ, Mirra SS, Binder LI, HansenLA, Wood JG. 1986. Filamentous aggre-gates in Pick’s disease, progressive supranu-clear palsy, and Alzheimer’s disease shareantigenic determinants with microtubule-associated protein, tau.Lancet 2(8517):1211

Poorkaj P, Bird TD, Wijsman E, Nemens E, Gar-ruto RM, et al. 1998. Tau is a candidate genefor chromosome 17 frontotemporal demen-tia. Ann. Neurol.43(6):815–25

Probst A, Gotz J, Wiederhold KH, TolnayM, Mistl C, et al. 2000. Axonopathy andamyotrophy in mice transgenic for humanfour-repeat tau protein.Acta Neuropathol.99(5):469–81

Probst A, Tolnay M, Langui D, Goedert M,Spillantini MG. 1996. Pick’s disease: hy-perphosphorylated tau protein segregates tothe somatoaxonal compartment.Acta Neu-ropathol.92(6):588–96

Prusiner SB. 1998.Prions. Proc. Natl. Acad.Sci. USA95(23):13363–83

Rebeiz J, Kolodny EH, Richardson EP Jr. 1967.Corticodentatonigral degeneration with neu-ronal achromasia: a progressive disorder inlate adult life. Trans. Am. Neurol. Assoc.92:23–26

Rebeiz JJ, Kolodny EH, Richardson EPJr. 1968. Corticodentatonigral degenerationwith neuronal achromasia 2.Arch. Neurol.18(1):20–33

Reed LA, Schmidt ML, Wszolek ZK, Balin BJ,Soontornniyomkij V, et al. 1998. The neu-ropathology of a chromosome 17-linked au-tosomal dominant parkinsonism and demen-tia (“pallido-ponto-nigral degeneration”).J.Neuropathol. Exp. Neurol.57(6):588–601

Rep. Work Group Frontotemporal DementiaPick’s Disease. 2001. Clinical and neu-ropathological diagnosis of frontotemporaldementias.Neurobiol. Aging.In press

Reynolds CH, Nebreda AR, Gibb GM, UttonMA, Anderton BH. 1997a. Reactivating ki-nase/p38 phosphorylates tau protein in vitro.J. Neurochem.69(1):191–98

Reynolds CH, Utton MA, Gibb GM, Yates A,Anderton BH. 1997b. Stress-activated pro-tein kinase/c-jun N-terminal kinase phos-phorylates tau protein.J. Neurochem.68(4):1736–44

Rizzini C, Goedert M, Hodges JR, SmithMJ, Jakes R, et al. 2000. Tau gene muta-tion K257T causes a tauopathy similar toPick’s disease.J. Neuropathol. Exp. Neurol.59(11):990–1001

Rizzu P, Joose M, Ravid R, Hoogeveen A, Kam-phorst W, et al. 2000. Mutation-dependentaggregation of tau protein and its selectivedepletion from the soluble fraction in brain ofP301L FTDP-17 patients.Hum. Mol. Genet.9(20):3075–82

Rizzu P, Van Swieten JC, Joosse M, HasegawaM, Stevens M, et al. 1999. High prevalence ofmutations in the microtubule-associated pro-tein tau in a population study of frontotempo-ral dementia in the Netherlands.Am. J. Hum.Genet.64(2):414–21

Ann

u. R

ev. N

euro

sci.

2001

.24:

1121

-115

9. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by O

pen

Uni

vers

ity o

n 07

/11/

13. F

or p

erso

nal u

se o

nly.

Page 36: Virginia M-Y Lee,1 Michel Goedert,2 and John Q Trojanowski

P1: FUM

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

1156 LEE ¥ GOEDERT ¥ TROJANOWSKI

Roy S, Datta CK, Hirano A, Ghatak NR, Zim-merman HM. 1974. Electron microscopicstudy of neurofibrillary tangles in Steele-Richardson-Olszewski syndrome.Acta Neu-ropathol.29(2):175–79

Sahara N, Tomiyama T, Mori H. 2000. Mis-sense point mutations of tau to segregate withFTDP-17 exhibit site-specific effects on mi-crotubule structure in COS cells: a novel ac-tion of R406W mutation.J. Neurosci. Res.60(3):380–87

Schmidt ML, Huang R, Martin JA, Henley J,Mawal-Dewan M, et al. 1996. Neurofibril-lary tangles in progressive supranuclear palsycontain the same tau epitopes identified inAlzheimer’s disease PHFtau.J. Neuropathol.Exp. Neurol.55(5):534–39

Schneider A, Biernat J, von Bergen M,Mandelkow E, Mandelkow EM. 1999.Phosphorylation that detaches tau pro-tein from microtubules (Ser262, Ser214)also protects it against aggregation intoAlzheimer paired helical filaments.Biochem-istry 38(12):3549–58

Schweers O, Sch¨onbrunn-Hanebeck E, MarxA, Mandelkow E. 1994. Structural studies oftau protein and Alzheimer paired helical fil-aments show no evidence for beta-structure.J. Biol. Chem.269(39):24290–97

Sengupta A, Wu Q, Grundke-Iqbal I, IqbalK, Singh TJ. 1997. Potentiation of GSK-3-catalyzed Alzheimer-like phosphorylationof human tau by cdk5.Mol. Cell Biochem.167(1–2):99–105

Sergeant N, David JP, Lefranc D, Vermersch P,Wattez A, Delacourte A. 1997. Different dis-tribution of phosphorylated tau protein iso-forms in Alzheimer’s and Pick’s diseases.FEBS Lett.412(3):578–82

Sergeant N, Wattez A, Delacourte A. 1999.Neurofibrillary degeneration in progressivesupranuclear palsy and corticobasal degener-ation: tau pathologies with exclusively “exon10” isoforms.J. Neurochem.72(3):1243–49

Seubert P, Mawal-Dewan M, Barbour R, JakesR, Goedert M, et al. 1995. Detection of phos-phorylated Ser262 in fetal tau, adult tau, and

paired helical filament tau.J. Biol. Chem.270(32):18917–22

Shin RW, Iwaki T, Kitamoto T, Tateishi J. 1991.Hydrated autoclave pretreatment enhancestau immunoreactivity in formalin-fixed nor-mal and Alzheimer’s disease brain tissues.Lab. Invest.64(5):693–702

Si ZH, Rauch D, Stoltzfus CM. 1998. Theexon splicing silencer in human immunode-ficiency virus type 1 Tat exon 3 is bipartiteand acts early in spliceosome assembly.Mol.Cell Biol. 18(9):5404–13

Singh TJ, Zaidi T, Grundke-Iqbal I, Iqbal K.1995. Modulation of GSK-3-catalyzed phos-phorylation of microtubule-associated pro-tein tau by nonproline-dependent protein ki-nases.FEBS Lett.358(1):4–8

Snow AD, Mar H, Nochlin D, Sekiguchi RT,Kimata K, et al. 1990. Early accumulationof heparan sulfate in neurons and in thebeta-amyloid protein-containing lesions ofAlzheimer’s disease and Down’s syndrome.Am. J. Pathol.137(5):1253–70

Sobue K, Agarwal-Mawal A, Wei L, Sun W,Miura Y, Paudel HK. 2000. Interaction ofneuronal cdc2 like protein kinase with micro-tubule associated protein tau.J. Biol. Chem.275(22):16673–80

Sontag E, Nunbhakdi-Craig V, Bloom GS,Mumby MC. 1995. A novel pool of pro-tein phosphatase 2A is associated with mi-crotubules and is regulated during the cellcycle.J. Cell Biol.128(6):1131–44

Sontag E, Nunbhakdi-Craig V, Lee G, BrandtR, Kamibayashi C, et al. 1999. Molecularinteractions among protein phosphatase 2A,tau, and microtubules. Implications for theregulation of tau phosphorylation and thedevelopment of tauopathies.J. Biol. Chem.274(36):25490–98

Sperfeld AD, Collatz MB, Baier H, Palm-bach M, Storch A, et al. 1999. FTDP-17:an early-onset phenotype with parkinsonismand epileptic seizures caused by a novel mu-tation.Ann. Neurol.46(5):708–15

Spillantini MG, Bird TD, Ghetti B. 1998a.Frontotemporal dementia and parkinsonism

Ann

u. R

ev. N

euro

sci.

2001

.24:

1121

-115

9. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by O

pen

Uni

vers

ity o

n 07

/11/

13. F

or p

erso

nal u

se o

nly.

Page 37: Virginia M-Y Lee,1 Michel Goedert,2 and John Q Trojanowski

P1: FUM

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

NEW INSIGHTS INTO TAUOPATHIES 1157

linked to chromosome 17: a new group oftauopathies.Brain Pathol.8(2):387–402

Spillantini MG, Crowther RA, Goedert M.1996. Comparison of the neurofibrillarypathology in Alzheimer’s disease and fa-milial presenile dementia with tangles.ActaNeuropathol.92(1):42–48

Spillantini MG, Crowther RA, Kamphorst W,Heutink P, Van Swieten JC. 1998b. Taupathology in two Dutch families with mu-tations in the microtubule-binding region oftau.Am. J. Pathol.153(5):1359–63

Spillantini MG, Goedert M, Crowther RA, Mur-rell JR, Farlow MR, Ghetti B. 1997. Famil-ial multiple system tauopathy with preseniledementia: a disease with abundant neuronaland glial tau filaments.Proc. Natl. Acad. Sci.USA94(8):4113–18

Spillantini MG, Murrell JR, Goedert M, Far-low MR, Klug A, Ghetti B. 1998c. Mu-tation in the tau gene in familial multiplesystem tauopathy with presenile dementia.Proc. Natl. Acad. Sci. USA95(13):7737–41

Spillantini MG, Yoshida H, Rizzini C, LantosPL, Khan N, et al. 2000. A novel tau mutation(N296N) in familial dementia with swollenachromatic neurons and corticobasal inclu-sion bodies.Ann. Neurol.48(6):939–43

Spittaels K, Van den Haute C, Van Dorpe J,Bruynseels K, Vandezande K, et al. 1999.Prominent axonopathy in the brain and spinalcord of transgenic mice overexpressing four-repeat human tau protein.Am. J. Pathol.155(6):2153–65

Spittaels K, Van den Haute C, Van Dorpe J,Geerts H, Mercken M, et al. 2000. Glyco-gen synthase kinase-3beta phosphorylatesprotein tau and rescues the axonopathy inthe central nervous system of human four-repeat tau transgenic mice.J. Biol. Chem.275(52):41340–49

Stanford PM, Halliday GM, Brooks WS, KwokJB, Storey CE, et al. 2000. Progressivesupranuclear palsy pathology caused by anovel silent mutation in exon 10 of thetau gene: expansion of the disease pheno-

type caused by tau gene mutations.Brain123(5):880–93

Steele JC, Richardson JC, Olszewski J. 1964.Progressive supranuclear palsy.Arch. Neu-rol. 10:333–59

Sturchler-Pierrat C, Abramowski D, DukeM, Wiederhold KH, Mistl C, et al. 1997.Two amyloid precursor protein transgenicmouse models with Alzheimer disease-like pathology.Proc. Natl. Acad. Sci. USA94(24):13287–92

Takahashi M, Tomizawa K, Ishiguro K, Taka-matsu M, Fujita SC, Imahori K. 1995. In-volvement of tau protein kinase I in pairedhelical filament-like phosphorylation of thejuvenile tau in rat brain.J. Neurochem.64(4):1759–68

Takahashi M, Yasutake K, Tomizawa K. 1999.Lithium inhibits neurite growth and tau pro-tein kinase I/glycogen synthase kinase-3β-dependent phosphorylation of juvenile tauin cultured hippocampal neurons.J. Neu-rochem.73(5):2073–83

Tanaka K, Watakabe A, Shimura Y. 1994. Poly-purine sequences within a downstream exonfunction as a splicing enhancer.Mol. CellBiol. 14(2):1347–54

Tellez-Nagel I, Wisniewski HM. 1973. Ul-trastructure of neurofibrillary tangles inSteele-Richardson-Olszewski syndrome.Arch. Neurol.29(5):324–27

Tesseur I, Van Dorpe J, Spittaels K, Van denHaute C, Moechars D, Van Leuven F. 2000.Expression of human apolipoprotein E4 inneurons causes hyperphosphorylation of pro-tein tau in the brains of transgenic mice.Am.J. Pathol.156(3):951–64

Tolnay M, Spillantini MG, Rizzini C, EcclesD, Lowe J, Ellison D. 2000. A new case offrontotemporal dementia and parkinsonismresulting from an intron 10+3-splice sitemutation in the tau gene: clinical and patho-logical features.Neuropathol. Appl. Neuro-biol. 26(4):368–78

Trojanowski JQ, Goedert M, Iwatsubo T,Lee VM-Y. 1998. Fatal attraction: abnor-mal protein aggregation and neuron death in

Ann

u. R

ev. N

euro

sci.

2001

.24:

1121

-115

9. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by O

pen

Uni

vers

ity o

n 07

/11/

13. F

or p

erso

nal u

se o

nly.

Page 38: Virginia M-Y Lee,1 Michel Goedert,2 and John Q Trojanowski

P1: FUM

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

1158 LEE ¥ GOEDERT ¥ TROJANOWSKI

Parkinson’s disease and Lewy body demen-tia. Cell Death Differ.5:832–37

Trojanowski JQ, Lee VM-Y. 1994. Pairedhelical filament tau in Alzheimer’s dis-ease. The kinase connection.Am. J. Pathol.144(3):449–53

Tsai LH, Delalle I, Caviness VS Jr, Chae T,Harlow E. 1994. p35 is a neural-specific reg-ulatory subunit of cyclin-dependent kinase 5.Nature371(6496):419–23

Tseng HC, Graves DJ. 1998. Natural methyl-amine osmolytes, trimethylamine N-oxideand betaine, increase tau-induced polymer-ization of microtubules.Biochem. Biophys.Res. Commun.250(3):726–30

Tseng HC, Lu Q, Henderson E, Graves DJ.1999. Phosphorylated tau can promote tubu-lin assembly.Proc. Natl. Acad. Sci. USA96(17):9503–8

Van Swieten JC, Stevens M, Rosso SM, Rizzu P,Joosse M, et al. 1999. Phenotypic variationin hereditary frontotemporal dementia withtau mutations.Ann. Neurol.46(4):617–26

Varani L, Hasegawa M, Spillantini MG, SmithMJ, Murrell JR, et al. 1999. Structure oftau exon 10 splicing regulatory elementRNA and destabilization by mutations offrontotemporal dementia and parkinsonismlinked to chromosome 17.Proc. Natl. Acad.Sci. USA96(14):8229–34

Varani L, Spillantini MG, Goedert M, Varani G.2000. Structural basis for recognition of theRNA major groove in the tau exon 10 splic-ing regulatory element by aminoglycosideantibiotics.Nucleic Acids Res.28(3):710–19

Verbeek MM, Otte-Holler I, van den Born J, vanden Heuvel LP, David G, et al. 1999. Agrinis a major heparan sulfate proteoglycan accu-mulating in Alzheimer’s disease brain.Am. J.Pathol.155(6):2115–25

Vermersch P, Robitaille Y, Bernier L, WattezA, Gauvreau D, Delacourte A. 1994. Bio-chemical mapping of neurofibrillary degen-eration in a case of progressive supranuclearpalsy: evidence for general cortical involve-ment.Acta Neuropathol.87(6):572–77

Vogelsberg-Ragaglia V, Bruce J, Richter-

Lansberg C, Zhang B, Hong M, et al. 2000.Distinct FTDP-17 missense mutations in tauproduce tau aggregates and other phatholo-gical phenotypes in transfected CHO cells.Mol. Biol. Cell.11(12):4093–104

von Bergen M, Friedhoff P, Biernat J, HeberleJ, Mandelkow EM, Mandelkow E. 2000. As-sembly of tau protein into Alzheimer pairedhelical filaments depends on a local se-quence motif ((306)VQIVYK(311)) formingbeta structure.Proc. Natl. Acad. Sci. USA97(10):5129–34

Watanabe A, Hasegawa M, Suzuki M, TakioK, Morishima-Kawashima M, et al. 1993. Invivo phosphorylation sites in fetal and adultrat tau.J. Biol. Chem.268(34):25712–17

Weingarten MD, Lockwood AH, Hwo SY,Kirschner MW. 1975. A protein factor es-sential for microtubule assembly.Proc. Natl.Acad. Sci. USA72(5):1858–62

Wijker M, Wszolek ZK, Wolters EC, RooimansMA, Pals G, et al. 1996. Localization ofthe gene for rapidly progressive autosomaldominant parkinsonism and dementia withpallido-ponto-nigral degeneration to chro-mosome 17q21.Hum. Mol. Genet.5(1):151–54

Wilhelmsen KC, Lynch T, Pavlou E, Hig-gins M, Nygaard TG. 1994. Localizationof disinhibition-dementia-parkinsonism-amyotrophy complex to 17q21–22.Am. J.Hum. Genet.55(6):1159–65

Wille H, Drewes G, Biernat J, Mandelkow EM,Mandelkow E. 1992. Alzheimer-like pairedhelical filaments and antiparallel dimersformed from microtubule-associated proteintau in vitro.J. Cell Biol.118(3):573–84

Wilson DM, Binder LI. 1997. Free fattyacids stimulate the polymerization of tauand amyloid beta peptides. In vitro evi-dence for a common effector of pathogen-esis in Alzheimer’s disease.Am. J. Pathol.150(6):2181–95

Wischik CM, Novak M, Thogersen HC, Ed-wards PC, Runswick MJ, et al. 1988. Iso-lation of a fragment of tau derived fromthe core of the paired helical filament of

Ann

u. R

ev. N

euro

sci.

2001

.24:

1121

-115

9. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by O

pen

Uni

vers

ity o

n 07

/11/

13. F

or p

erso

nal u

se o

nly.

Page 39: Virginia M-Y Lee,1 Michel Goedert,2 and John Q Trojanowski

P1: FUM

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

NEW INSIGHTS INTO TAUOPATHIES 1159

Alzheimer disease.Proc. Natl. Acad. Sci.USA85(12):4506–10

Woody RW, Clark DC, Roberts GC, Martin SR,Bayley PM. 1983. Molecular flexibility inmicrotubule proteins: proton nuclear mag-netic resonance characterization.Biochem-istry 22(9):2186–92

Yamada T, McGeer PL, McGeer EG. 1992.Appearance of paired nucleated, tau-positiveglia in patients with progressive supranuclearpalsy brain tissue.Neurosci. Lett.135(1):99–102

Yamaguchi H, Ishiguro K, Uchida T, TakashimaA, Lemere CA, Imahori K. 1996. Preferentiallabeling of Alzheimer neurofibrillary tangleswith antisera for tau protein kinase (TPK)I/glycogen synthase kinase-3 beta and cyclin-dependent kinase 5, a component of TPK II.Acta Neuropathol.92(3):232–41

Yasuda M, Kawamata T, Komure O, KunoS, D’Souza I, et al. 1999. A mutation inthe microtubule-associated protein tau inpallido-nigro-luysian degeneration.Neurol-ogy53(4):864–68

Yasuda M, Takamatsu J, D’Souza I, CrowtherRA, Kawamata T, et al. 2000. A novel mu-

tation at posititon+12 in the intron fol-lowing exon 10 of the tau gene in familialfrontotemporal dementia (FTD-Kumamoto).Ann. Neurol.47(4):422–29

Yen SH, Hutton M, DeTure M, Ko LW,Nacharaju P. 1999. Fibrillogenesis of tau: in-sights from tau missense mutations in FTDP-17.Brain Pathol.9(4):695–705

Yoshida H, Ihara Y. 1993. Tau in paired helicalfilaments is functionally distinct from fetaltau: assembly incompetence of paired helicalfilament-tau.J. Neurochem.61(3):1183–86

Zheng-Fischh¨ofer Q, Biernat J, MandelkowEM, Illenberger S, Godemann R, MandelkowE. 1998. Sequential phosphorylation of tauby glycogen synthase kinase-3β and proteinkinase A at Thr212 and Ser214 gener-ates the Alzheimer-specific epitope of anti-body AT100 and requires a paired-helical-filament-like conformation.Eur. J. Biochem.252(3):542–52

Zhukareva V, Vogelsberg-Ragaglia V, VanDeerlin VMD, Bruce J, Schuck T, et al. 2001.Loss of brain tau defines novel sporadic andfamilial tauopathies with frontotemporal de-mentia.Ann. Neurol.49(1):165–75

Ann

u. R

ev. N

euro

sci.

2001

.24:

1121

-115

9. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by O

pen

Uni

vers

ity o

n 07

/11/

13. F

or p

erso

nal u

se o

nly.

Page 40: Virginia M-Y Lee,1 Michel Goedert,2 and John Q Trojanowski

Annual Review of Neuroscience Volume 24, 2001

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

THE ROLE AND REGULATION OF ADENOSINE IN THE CENTRAL NERVOUS SYSTEM, Thomas V. Dunwiddie, Susan A. Masino

31

LOCALIZATION AND GLOBALIZATION IN CONSCIOUS VISION, S. Zeki 57

GLIAL CONTROL OF NEURONAL DEVELOPMENT, Greg Lemke 87TOUCH AND GO: Decision-Making Mechanisms in Somatosensation, Ranulfo Romo, Emilio Salinas 107

SYNAPTIC MODIFICATION BY CORRELATED ACTIVITY: Hebb''s Postulate Revisited, Guo-qiang Bi, Mu-ming Poo 139

AN INTEGRATIVE THEORY OF PREFRONTAL CORTEX FUNCTION, Earl K. Miller, Jonathan D. Cohen 167

THE PHYSIOLOGY OF STEREOPSIS, B. G. Cumming, G. C. DeAngelis 203

PARANEOPLASTIC NEUROLOGIC DISEASE ANTIGENS: RNA-Binding Proteins and Signaling Proteins in Neuronal Degeneration, Kiran Musunuru, Robert B. Darnell

239

ODOR ENCODING AS AN ACTIVE, DYNAMICAL PROCESS: Experiments, Computation, and Theory, Gilles Laurent, Mark Stopfer, Rainer W Friedrich, Misha I Rabinovich, Alexander Volkovskii, Henry DI Abarbanel

263

PROTEIN SYNTHESIS AT SYNAPTIC SITES ON DENDRITES, Oswald Steward, Erin M. Schuman 299

SIGNALING AND TRANSCRIPTIONAL MECHANISMS IN PITUITARY DEVELOPMENT, Jeremy S. Dasen, Michael G. Rosenfeld

327

NEUROPEPTIDES AND THE INTEGRATION OF MOTOR RESPONSES TO DEHYDRATION , Alan G. Watts 357

THE DEVELOPMENTAL BIOLOGY OF BRAIN TUMORS, Robert Wechsler-Reya, Matthew P. Scott 385

TO EAT OR TO SLEEP? OREXIN IN THE REGULATION OF FEEDING AND WAKEFULNESS, Jon T. Willie, Richard M. Chemelli, Christopher M. Sinton, Masashi Yanagisawa

429

SPATIAL PROCESSING IN THE BRAIN: The Activity of Hippocampal Place Cells, Phillip J. Best, Aaron M. White, Ali Minai 459

THE VANILLOID RECEPTOR: A Molecular Gateway to the Pain Pathway, Michael J Caterina, David Julius 487

PRION DISEASES OF HUMANS AND ANIMALS: Their Causes and Molecular Basis, John Collinge 519

VIKTOR HAMBURGER AND RITA LEVI-MONTALCINI: The Path to the Discovery of Nerve Growth Factor, W. Maxwell Cowan 551

EARLY DAYS OF THE NERVE GROWTH FACTOR PROTEINS, Eric M. Shooter 601

SEQUENTIAL ORGANIZATION OF MULTIPLE MOVEMENTS: Involvement of Cortical Motor Areas, Jun Tanji 631

INFLUENCE OF DENDRITIC CONDUCTANCES ON THE INPUT-OUTPUT PROPERTIES OF NEURONS, Alex Reyes 653

Ann

u. R

ev. N

euro

sci.

2001

.24:

1121

-115

9. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by O

pen

Uni

vers

ity o

n 07

/11/

13. F

or p

erso

nal u

se o

nly.

Page 41: Virginia M-Y Lee,1 Michel Goedert,2 and John Q Trojanowski

NEUROTROPHINS: Roles in Neuronal Development and Function, Eric J Huang, Louis F Reichardt 677

CONTRIBUTIONS OF THE MEDULLARY RAPHE AND VENTROMEDIAL RETICULAR REGION TO PAIN MODULATION AND OTHER HOMEOSTATIC FUNCTIONS, Peggy Mason

737

ACTIVATION, DEACTIVATION, AND ADAPTATION IN VERTEBRATE PHOTORECEPTOR CELLS, Marie E Burns, Denis A Baylor 779ACTIVITY-DEPENDENT SPINAL CORD PLASTICITY IN HEALTH AND DISEASE, Jonathan R Wolpaw, Ann M Tennissen 807QUANTITATIVE GENETICS AND MOUSE BEHAVIOR, Jeanne M Wehner, Richard A Radcliffe, Barbara J Bowers 845

EARLY ANTERIOR/POSTERIOR PATTERNING OF THE MIDBRAIN AND CEREBELLUM, Aimin Liu, Alexandra L Joyner 869

NEUROBIOLOGY OF PAVLOVIAN FEAR CONDITIONING, Stephen Maren 897

{{alpha}}-LATROTOXIN AND ITS RECEPTORS: Neurexins and CIRL/Latrophilins, Thomas C Südhof 933IMAGING AND CODING IN THE OLFACTORY SYSTEM, John S Kauer, Joel White 963

THE ROLE OF THE CEREBELLUM IN VOLUNTARY EYE MOVEMENTS, Farrel R Robinson, Albert F Fuchs 981

ROLE OF THE REELIN SIGNALING PATHWAY IN CENTRAL NERVOUS SYSTEM DEVELOPMENT, Dennis S Rice, Tom Curran 1005

HUMAN BRAIN MALFORMATIONS AND THEIR LESSONS FOR NEURONAL MIGRATION, M Elizabeth Ross, Christopher A Walsh 1041

MORPHOLOGICAL CHANGES IN DENDRITIC SPINES ASSOCIATED WITH LONG-TERM SYNAPTIC PLASTICITY, Rafael Yuste, Tobias Bonhoeffer

1071

STOPPING TIME: The Genetics of Fly and Mouse Circadian Clocks, Ravi Allada, Patrick Emery, Joseph S. Takahashi, Michael Rosbash

1091

NEURODEGENERATIVE TAUOPATHIES, Virginia M-Y Lee, Michel Goedert, John Q Trojanowski 1121

MATERNAL CARE, GENE EXPRESSION, AND THE TRANSMISSION OF INDIVIDUAL DIFFERENCES IN STRESS REACTIVITY ACROSS GENERATIONS, Michael J Meaney

1161

NATURAL IMAGE STATISTICS AND NEURAL REPRESENTATION, Eero P Simoncelli, Bruno A Olshausen 1193

Nerve Growth Factor Signaling, Neuroprotection, and Neural Repair, Michael V Sofroniew, Charles L Howe, William C Mobley 1217

FLIES, GENES, AND LEARNING, Scott Waddell, William G Quinn 1283

Ann

u. R

ev. N

euro

sci.

2001

.24:

1121

-115

9. D

ownl

oade

d fr

om w

ww

.ann

ualr

evie

ws.

org

by O

pen

Uni

vers

ity o

n 07

/11/

13. F

or p

erso

nal u

se o

nly.