You are on page 1of 15

NeuroMolecular Medicine

Copyright © 2002 Humana Press Inc.


All rights of any nature whatsoever reserved.
ISSN1535-1084/02/02:151–165/$20.00

Axonal Transport, Tau Protein, and Neurodegeneration


in Alzheimer’s Disease

Dick Terwel, Ilse Dewachter, and Fred Van Leuven*


Experimental Genetics Group, K.U.Leuven, B-3000 Leuven, Belgium
Received May 21, 2002; Accepted May 22, 2002

Abstract
The molecular causes and the genetic and environmental modifying factors of the sporadic
form of Alzheimer’s disease (AD) remain elusive. Extrapolating from the known mutations that
cause the rare familial forms and from the typical post-mortem pathological lesions in all AD
patients—e.g., amyloid plaques and neurofibrillary tangles (NFTs)—the evident molecular can-
didates are amyloid precursor protein (APP), presenilin, and tau protein. To include ApoE4 as
the only certain genetic modifier known leaves us to face the challenge of implementing these
very different molecules into an evident pathological partnership. In more than one respect,
the proposition of disturbed axonal transport appears attractive with more details becoming
available on APP processing and microtubular transport and also of the pathology in the model
systems—e.g., transgenic mice expressing APP or protein tau. Conversely, the resistance of APP-
transgenic mice with full-blown amyloid pathology to also develop tau-related neurofibrillar
pathology is a major challenge for this hypothesis. From the most relevant data discussed here,
we conclude that the postulate of disturbed axonal transport as the primary event in AD is dif-
ficult to defend. On the other hand, failing axonal transport appears to be of major importance
in the later stages in AD, by further compromising tau protein, APP metabolism, and synaptic
functioning. Protein tau may thus be the central “executer” in the chain of events leading from
amyloid neurotoxicity to tau hyperphosphorylation, microtubular destabilization, disturbed
axonal transport, and synaptic failure to neurodegeneration. In order to identify normal phys-
iological processes and novel pathological targets, definition is needed—in molecular detail—
of the complex mechanisms involved.

Index Entries: axonal transport; Alzheimer’s disease; transgenic mice; protein tau; GSK-3β;
microtubules; amyloid.

*Authors to whom all correspondence and reprint requests should be addressed. E-mail: fredvl@med.kuleuven.ac.be;
www.med.kuleuven.ac.be/legtegg/

NeuroMolecular Medicine 151 Volume 2, 2002


152 Terwel et al.

Introduction other hand, recent findings propose a direct role for


APP in axonal transport, as APP can link to kinesins
The Problem of Plaques and Tangles moving along the microtubules (Kamal et al., 2001).
This evidently brings many factors involved in AD
In patients suffering from Alzheimer’s disease in close proximity to each other, and the findings
(AD), the brain is histopathologically characterized may alter our perspective of the role of axonal trans-
by the presence of extracellular plaques made up of port in AD, which is the topic of this article.
β-amyloid peptides (Aβs) and intraneuronal fibril- Although numerous reviews have been dedicated
lary tangles made up of aggregated tau protein. The to the amyloid aspect of the disease, this article
direct or indirect mechanistical relationship between focuses on the pathological role of tau and related
tangles and plaques remains elusive. mechanisms, as studied in our transgenic mice. We
In a small percentage of AD cases (less than 1%), place these findings into the context of recent find-
the disease is strictly and dominantly inherited ings that might link both sides of the medal, and
through mutations in the genes that code for amy- may even incorporate the mysterious role of ApoE4.
loid precursor protein (APP) or for one of the pre-
senilins. The Aβs are liberated from APP by the Tau Protein: Structure and Function
action of two distinct proteinases—e.g., the first
cleaving at the N-terminal side of the amyloid Tau protein is predominantly expressed in neu-
sequence, known as the β-site of APP, giving rise to rons of both the central and peripheral nervous
the names β-secretase or BACE, and the second system. It is encoded by a single gene with 16 exons
named γ-secretase, an activity closely involving pre- located on the long arm of chromosome 17 (for
senilins (for reviews see Esler and Wolfe, 2001; Siso- review see Buée et al., 2000). By alternative splicing,
dia and St George-Hyslop, 2002). Whether presenilin the primary transcript can produce up to six dif-
is molecularly identical to γ-secretase or an essen- ferent mRNAspecies in the brain, coding for six tau-
tial associated cofactor is a matter of debate. Nev- protein isoforms ranging from 352 to 441 amino
ertheless, all known mutations in APP and in acids. These six isoforms contain either three or four
presenilins that cause early-onset familial AD (FAD) imperfect C-terminal tandem repeats (TRs) and
increase either the total levels of Aβ or the ratio of either one or two shorter N-terminal domains (N).
Aβ42/Aβ40 in the brain, thereby tilting the balance The shortest tau-protein isoform is the only one pre-
toward amyloid deposits. sent in the fetal human brain, and all six isoforms
No mutations that cause AD have been described are present in the adult human brain.
in the gene that codes for tau protein (chromosome An important function of tau protein involves the
17). Conversely, mutations in the coding region of stabilization and spacing of microtubules, proba-
tau protein or in intron 10 of the tau gene are associ- bly by linking to a number of tubulin subunits,
ated with a group of diseases known as frontotem- thereby preventing or slowing microtubule depoly-
poral dementia (FTD) with parkinsonism linked to merization (Drubin and Kirschner, 1986). The three
chromosome 17 (FTDP-17), presenting with abnor- or four C-terminal repeats that mediate binding of
mal tau aggregates only (reviewed by Delacourte and tau protein to the microtubules (Lee et al., 1989) con-
Buée, 2000). Depending on the particular mutation tain highly conserved 18-amino-acid stretches sepa-
in the tau protein, the tau filaments formed in neu- rated by less conserved 13- or 14-amino acid
rons in the brains of these patients can be either inter-repeat domains (Goedert et al., 1989). The clus-
straight, twisted or paired helical filaments (PHFs). ter of three or four microtubule-binding domains are
However, the fact that mutant tau proteins form N-terminally flanked by a proline-rich region and
aggregates and are the primary cause of these neu- C-terminally by the so-called 5th repeat (also named
rodegenerative diseases has sparked renewed inter- R′). Although the three or four C-terminal repeats are
est into the role of tau protein in normal physiology essential for effective binding of tau protein to the
and in the pathology of AD. microtubules, the affinity of binding is further
Tau protein is a typical microtubule-associated increased by their flanking regions. This model or
protein (MAP) and thus is directly implicated in mechanism is known as the “jaws-model”: the cen-
maintaining the integrity and stability of the micro- tral repeats enable rapid binding to microtubules,
tubules and involved in axonal transport. On the and the flanking regions are considered to be

NeuroMolecular Medicine Volume 2, 2002


Axonal Transport in Alzheimer’s Disease 153

“embracing” domains, augmenting the affinity (Man- phorylation sites, and at least 30 of these were effec-
delkow et al., 1995). It is to be remembered that tau tively identified experimentally as being phospho-
protein with four C-terminal domains (tau-4R) binds rylated in one system or another (reviewed by Buée
more tightly to microtubules than three-repeat tau et al. 2000). Many phosphorylation sites are serine
(tau-3R) (Goedert and Jakes, 1990; Scott et al., 1991). or threonine followed by proline, and most are
The microtubule-binding properties of tau protein located outside the microtubule-binding repeat
are believed to be important for a number of region. Among the specific “proline-dependent”
processes, e.g., the formation and maintenance of kinases that can phosphorylate protein tau at dif-
axons and for fast axonal transport (FAT). Down- ferent sites in vitro, special attention is dedicated to
regulation of expression of tau protein in primary glycogen-synthase kinase-3β (GSK-3β) (Michel et
cerebellar neurons with anti-sense oligonucleotides al., 1998), mitogen-activated protein kinase (MAPK)
decreases their ability to generate axons (Caceras and (Drewes et al., 1992), stress-activated protein kinases
Kosik, 1990; Caceras et al., 1991). Similarly, tau anti- (SAPKs) (Goedert et al., 1997) and cyclin-dependent
sense oligonucleotides produces retraction of neu- kinases (CDKs) including cdc2 and cdk5 (Baumann
rites in PC12 cells treated with nerve growth factor et al., 1993; Patrick et al., 1999).
(Hanemaaijer and Ginzburg, 1991). These observa- Other serine and threonine residues, not followed
tions suggest that protein tau is involved in assem- by proline, are phosphorylated by other protein
bling neuronal microtubules into the organized kinases, including microtubule-affinity-regulating
arrays required for the elaboration and maintenance kinase (MARK) (Drewes et al., 1993), calcium/
of axons. Injection of tau protein in Sf9 insect cells calmodulin kinase II (CAMKII), cAMP-dependent
has induced axon-like processes containing polar- kinase (PKA) (Johnson et al., 1992), and casein kinase
ized bundles of microtubules as found in axons (Baas II (Greenwood et al., 1994). Tau protein is rapidly
et al., 1991; Biernat and Mandelkow, 1999). By con- dephosphorylated by endogenous phosphatases
trast, protein tau-deficient mice display a very light such as protein phosphatases 1, 2A, and 2B (cal-
phenotype and only very modest alterations of their cineurin) that are all present in the brain, and effec-
microtubule organization in small-caliber axons. This tively dephosphorylate tau protein in vitro. Since
can be explained by the increased expression of endogenous, active tau protein is constitutively
microtubule-associated protein (MAP) 1A, that phosphorylated at many sites, and rather similar to
would compensate for the absent tau protein (Harada tau proteins isolated from PHF in the AD brain
et al., 1994). (Matsuo et al., 1994), it is accepted that phospho-
One disruptive effect or toxic gain function of tau rylation and dephosphorylation is the control mech-
protein in intracellular transport has become evident anism that regulates binding and dissociation of tau
from studies on stably transfected Chinese hamster protein to and from microtubules.
ovary (CHO) cells and differentiated neuroblastoma Protein tau is “hyper”-phosphorylated in the
(N2a) cells (Ebneth et al., 1998; Trinczek et al., 1999). brains of patients suffering from AD or from other
Special mention should be made of primary cultures neurodegenerative diseases characterized by neu-
of dorsal root ganglion (DRG) cells from our trans- ronal tau protein aggregates (Buée et al., 2000).
genic mice that overexpress 4R human tau (htau-4R). Apparently, this is why hyperphosphorylation of
In these cells, the overexpression of tau interfered protein tau has been considered important for its
with anterograde intracellular transport of cyto- disturbed function and for its self-aggregation,
plasmic organelles and intermediate filaments medi- although it is not clear whether the higher number
ated by kinesin-like motor proteins along of phosphate groups or the phosphorylation of spe-
microtubules (Nuydens et al., 2002). Transgenic mice cific sites is more important. It is also not clear
that overexpress tau and tau-kinases have been gen- whether the two phenomena—e.g., hyperphos-
erated, and have revealed relevant aspects of phorylation and aggregation—are inherently and
(dys)function of tau protein, as discussed further. functionally linked, or whether they are the result
of the same structural feature(s) of tau protein, as
Tau Phosphorylation and Tangle disturbed by phosphorylation. However, it is clear
Formation in AD that aggregation per se will prevent effective bind-
Tau protein is a phosphoprotein: the longest brain ing to microtubules, and effectively lowers the con-
isoform has 79 putative serine and threonine phos- centration of free, active tau protein. As far as we

NeuroMolecular Medicine Volume 2, 2002


154 Terwel et al.

know, aggregation of tau protein is not known to processes that cannot and need not be discussed
be a control mechanism in its own right. here.
Following the observation that increased phos- However, an even greater enigma remains—the
phorylation of tau protein is an early change in those relation of tau pathology to processing of APP into
brain areas where tangle pathology is manifested its metabolites, mainly the Aβs and C99 fragments.
in the AD brain (Braak et al., 1994; Delacourte et al., Indeed, it is totally uncertain whether the increased
1999) and because hyperphosphorylated tau pro- Aβ level, which is accepted by many as the primary
tein has a reduced affinity for microtubules (Iqbal, event in the pathology of AD, is in any way related
1986; Yoshida and Ihara, 1993) a model was pro- to increased phosphorylation of tau protein. The
posed to link both. Essentially, increased phospho- most intricate aspects are not examined here because
rylation of tau protein by different kinases is of limited space. We must consider two essential
believed to be the primary event(s) preventing options: (i) a co-effect, or the same triggers that
hyperphosphorylated tau to bind to the micro- derange APP processing also govern hyperphos-
tubules, causing their destabilization and interfer- phorylation of tau protein, or alternatively (ii) a
ing with axonal transport. It has been proposed that sequential process, whereby unknown causes lead
tau hyperphosphorylation results from oxidative to mis-processing of APP, which in turn deranges
stress and covalent modification of tau by the lipid the mechanisms that control tau protein. Evidently,
peroxidation product 4-hydroxynonenal (Mattson this neither grants—nor denies—hyperphosphory-
et al., 1997). The gradual increase of the cytoplas- lation of tau the role of executer!
mic levels of hyperphosphorylated tau in the The model of hyperphosphorylation and PHF
affected neurons progressively constitutes a pool of and NFT formation can only fit with deterioration
substrate from which the PHFs form and aggregate of axonal transport, if less active tau protein will
into neurofibrillary tangles (NFTs). effectively be available for stabilization of the micro-
tubules. Evidently, this occurs only when tran-
Causes and Consequences scriptional and translational activity and thereby
of Hyperphosphorylation of Tau Protein its synthesis remains unaltered by the post-
translational modifications—e.g., hyperphospho-
This model does not offer an explanation for the rylation of tau. This is a matter of debate, as it has
cause or for the identity of the increased kinase activ- not been unequivocally demonstrated in the brain
ity, and has also been challenged by the observa- of AD patients. In addition, however, hyperphos-
tion that phosphorylation of tau protein reduces its phorylated tau can sequester normal tau protein
aggregation in the presence of cofactors such as into filaments and thereby promote destabilization
heparin or other polyanions (Friedhoff et al., 2000). and disassembly of microtubules, as shown at least
However, in the absence of any cofactors, phos- in vitro (Alonso et al., 1996). Finally, the tau fila-
phorylation of tau protein was necessary and suf- ments and aggregates can form physical barriers or
ficient for its self-assembly into paired helical or otherwise interfere physically with anterograde and
straight filaments (Alonso et al., 2001). It is clear retrograde axonal transport, and thereby pose sec-
that the process of tau self-aggregation is not well- ondary problems.
understood, not even in vitro with recombinant tau
protein. The excess phosphate groups may simply Transgenic Mice with Tau
neutralize charges, shift properties of tau protein in
solution from a rather basic to a more acid protein, and Tau-Kinases
and thereby render it from a hydrophilic to a more Disruption of Axonal Transport
hydrophobic protein. Indeed, occupation of only
by Excess Tau
nine phosphorylation sites could be sufficient to
reduce or neutralize the basic character of tau pro- The remarkable effect of excess tau protein on
tein (Ruben et al., 1997). Thus, the conglomeration axonal transport has been originally exemplified in
of tau protein and other proteins into the massive vivo in our transgenic mice that overexpress the
NFTs, can be the result of subsequent modifications, longest four-repeat human tau protein isoform
such as the transglutamination and glycosylation (htau-2N, 4R) specifically in neurons (Spittaels et
(Norlund et al., 1999; Wang et al., 1996), which are al., 1999). These transgenic mice developed axonal

NeuroMolecular Medicine Volume 2, 2002


Axonal Transport in Alzheimer’s Disease 155

Fig. 1. Selected examples of axonal problems in the brain of tau-4R-transgenic mice. (A) Beginning dilatation
(arrow-heads) close to the cell body. (B) Overview of cortex showing numerous dilatations (arrow-heads). (C)
Dilatations of different size, revealed by silver staining. (D) Close-up view of a spheroid from panel B. (E) Ultra-
structural view of the contents of a spheroid: mitochondria (triangles), neurofilaments (small arrow-heads), micro-
tubule (small arrow), and unidentified vesicle (larger arrow). (A, B, D) the immunohistochemistry was performed
with antibody SMI32 directed against neurofilaments. Color image available for viewing at www.humanapress.com

degeneration in the brain and spinal cord that was that NFs contribute heavily to the axonopathy of
characterized by axonal dilations with accumula- tau transgenic mice (Ishihara et al., 2001a). How-
tion of neurofilaments (NFs), microtubules, mito- ever, problems with NFs are observed in AD only
chondria, and different types of vesicles (Fig. 1). in later stages, and a direct role of NFs and axonal
Subsequently, Wallerian degeneration and severe dilations in the early ontogeny of AD seems less
muscle wasting and motoric problems demon- likely.
strated the extensive neurodegeneration caused by Most recently, our tau transgenic mice have
the overexpression of human tau protein in a gene- proved to be relevant for prion-induced neurode-
dosage fashion. generation (Künzi et al., 2002). Tau transgenic mice
This phenotype is explained by the fact that the infected with prions accumulated intraneuronal
mouse thy-1 gene promoter did express tau protein deposits of hyperphosphorylated tau protein,
in all central neurons, including the motor neurons resembling Gerstmann-Sträussler-Scheinker syn-
in the spinal cord, which are known to be particu- drome. These findings identify tau pathology as a
larly sensitive to stress. In this respect, these find- possible end stretch of prion-induced neurodegen-
ings are of considerable interest in the field of eration (Künzi et al., 2002)
amyotrophic lateral sclerosis (ALS). The dilated For the sake of completeness, we also refer to tau-
axons or spheroids much resemble those observed 3R transgenic mice that developed another type of
in ALS and ALS/Parkinson dementia complex pathology in the hippocampus, e.g., straight fila-
(Nakazato et al., 1984; Shankar et al., 1989). In ALS, ments formed in aged mice older than 18 mo (Ishi-
accumulation of NFs is a prominent feature hara, 2001b), which was proposed to be relevant for
(Rouleau et al., 1996), and it has been demonstrated AD, given the age-dependence.

NeuroMolecular Medicine Volume 2, 2002


156 Terwel et al.

Specified mutations in the tau gene, including together with an increased accessibility of the
those in intron 10, cause neurodegeneration and J-domain motifs of the KLCs to HSC70, leading
have a remarkable molecular effect—only an to removal of kinesin from vesicles and their
increase in the levels of the functionally normal rapid degradation. Therefore, increased GSK-3β
tau-4R protein isoform (Hutton et al., 1998; Spillan- activity—and/or decreased de-phosphorylation—
tini et al., 1998; Hong et al., 1998; Hasegawa et al., can be anticipated to jeopardize FAT in two differ-
1998). Evidently, this argues for critical levels of ent ways.
protein tau-4R in the pathology of FTD and by exten- Direct evidence for a role of GSK-3β in tau
sion, in AD. As stated previously, we have unequiv- protein-related processes stems from our findings
ocally documented in the transgenic mice in vivo with single- and double-transgenic mice that over-
that elevated concentrations of tau protein blocked express htau-4R and/or GSK-3β (Spittaels et al.,
traffic of vesicles and organelles (Fig. 1) (Spittaels 1999, 2000). Neuronally overexpressed GSK-3β was
et al., 1999; 2000). demonstrated to hyperphosphorylate tau protein
The following proposition has been recently reit- in vivo in the brain and spinal cord of double-
erated that axonal transport in AD could become transgenic mice (Spittaels et al., 2000) thereby
disrupted by increased neuronal concentrations of reducing the amount of protein tau associated with
tau protein, now based on evidence of a cellular par- microtubules by 50% (Spittaels et al., 2000). More-
adigm, also demonstrating an effect on transport of over, the unbound tau protein was hyperphospho-
APP (Stamer et al., 2002). These results could help rylated and especially at the AD-2 epitope, e.g., an
to explain the increased Aβ production in the trans- epitope shown to contain serine 396 and serine 404.
Golgi network. The amount of Aβ produced could The region surrounding these serines in tau protein
be altered by delayed axonal transport, as well as is thus demonstrated to contain essential determi-
the precise species of metabolites of APP produced— nants governing tau protein—microtubule interac-
e.g., Aβ40 or 42, monomeric Aβ, or Aβ-oligomers or tion in vivo.
Aβ-derived diffusible ligands (ADDLs) (Lambert et Surprisingly, the axonopathy was rescued in the
al., 1998; Walsh et al., 2000). Moreover, the C99 frag- tau x GSK-3β double-transgenic mice, together
ments resulting from β-cleavage of APP and the with a near-total normalization of the functional
obligate precursors of the Aβs may be included disabilities. The explanation we proposed was
among the toxic metabolites of APP, as we have that increased phosphorylation of tau protein by
recently demonstrated in vivo in aged mice that the extra GSK-3β activity did effectively lower the
were neuronally deficient in PS1, and thereby in actual concentration of tau protein that was able
γ-secretase activity (Dewachter et al., 2002). to bind to the microtubules, and thereby reduced
Unfortunately, evidence for increased intracel- the interference with transport by the kinesin
lular tau concentrations in the brain of AD patients motors (Fig. 2). More recently, we observed that
is to be taken with caution, since it represents data the overexpression of GSK-3β on itself caused a
on total tau concentration, including deposited or light form of microcephaly in the GSK-3β-
“tangle” tau (Khatoon et al. 1992), while concen- transgenic mice. This appeared to be caused by a
trations of soluble tau in the AD brain have been decrease in the caliber of the neuronal processes
found to be reduced (Ksiezak-Reding et al., 1988). (Spittaels et al., 2002). Although much further work
is needed, these additional data demonstrate that
Rescue of Axonopathy in Tau Transgenic GSK-3β is intimately involved in the architecture
of axons and other neuronal processes, providing
Animals by Co-Expression of GSK-3β
indirect support for its role in tau-mediated con-
Vesicle stalling would fit well with the more clas- trol of axonal transport.
sical type of view on how hyperphosphorylation of The theory that phosphorylation of tau protein
tau protein causes problems with axonal transport. reduces binding to microtubules and restores axonal
In addition to a role in destabilization of micro- transport was further supported by studies in DRG
tubules (as discussed here), a role for GSK-3β has cells cultured from the transgenic mice that over-
been documented in regulating kinesin-based motil- express GSK-3β and htau (Nuydens et al., 2002).
ity (Morfini et al., 2002). Evidence that GSK-3β phos- DRG cells from htau-transgenic mice showed
phorylates kinesin light chain was presented reduced sprouting capacity, while density and sta-

NeuroMolecular Medicine Volume 2, 2002


Axonal Transport in Alzheimer’s Disease 157

Fig. 2. Schematic representation of impaired axonal transport by excess tau protein, and its rescue by
co-expression of GSK-3β. In essence, excess tau overexpressed in tau-4R-transgenic mice (e.g., tau in middle panel)
binds to microtubules and prevents kinesin motors to move, as they normally do (e.g., wild-type in upper panel).
Co-expression of GSK-3β phosphorylates tau protein, and thereby prevents it from binding to the microtubules
restoring normal axonal transport (lower panel). The schemes are based on extensive experimental data (Spittaels
et al., 1999, 2000, 2002), as discussed in the text. Color image available for viewing at www.humanapress.com

bility of microtubules in the axonal processes were et al., 1999; Van Dorpe et al., 2000). A role for an
increased. Video-enhanced contrast microscopy axonal-transport deficit in the development of
demonstrated a dramatic inhibition of FAT. Coex- pathology was not directly obvious, however
pression of GSK-3β increased tau protein phos- (Moechars et al., 1999; Van Dorpe et al., 2000;
phorylation and reversed the effects on microtubule Dewachter et al., 2000; and unpublished results).
stability and saltatory motion (Nuydens et al., 2002). Eventually, this could be explained by the lesser vul-
These studies demonstrate that GSK-3β is an nerability of mouse neurons to Aβs (Geula et al.,
effective tau-kinase in vivo, and that this phospho- 1998). Whether this is related to the fact that the
rylation is directly involved in the regulation of APP[V717I] mice fail to develop tangle pathology,
binding of tau protein to microtubules in vivo. Nev- despite the presence of typical tau epitopes in
ertheless, despite the high levels of hyperphos- swollen neurites surrounding the plaques
phorylated tau protein in the brain of the tau x (Moechars et al., 1999; Van Dorpe et al., 2000;
GSK-3β double-transgenic mice, no signs of Dewachter et al., 2000), is a distinct possibility.
PHF formation was evident in these studies, indi- In all transgenic mice that we generated to over-
cating that other factors are involved (Spittaels et express APP, tau protein-4R, GSK-3β, ApoE4, and
al., 1999, 2000). others, the transgene was controlled by the mouse
thy-1 gene promoter, which allows high expression
in all central neurons, including those of the spinal
Pathology in Mice that Co-Express cord (Moechars et al., 1999; Spittaels et al., 1999,
Tau Protein and APP 2000; Tesseur et al., 2000a,b). Therefore, we antici-
pated that if mere expression of APP in the spinal
If axonal transport is problematic in AD, then cord were to jeopardize axonal transport, that
alterations are also to be expected in APP transgenic would have been manifested by motoric problems,
mouse models. We have analyzed this aspect in our as seen in the tau-4R- and ApoE4-transgenic mice
APP[V717I]-transgenic mice, which neuronally (Spittaels et al., 1999, 2000; Tesseur et al., 2000a,b).
overexpress the London mutant of APP and thereby In sharp contrast to results obtained with overex-
recapitulate—robustly and progressively—the pression of APP in Drosophila larva (Gunawardena
amyloid pathology that is very reminiscent of AD and Goldstein, 2001) this theory was not shown to
patients, including vascular angiopathy (Moechars be accurate.

NeuroMolecular Medicine Volume 2, 2002


158 Terwel et al.

To investigate the synergism of APP and tau in into the fine details, mice deficient in APP have not
vivo, we have generated APP[V717I] x tau-4R helped very much to solve the mystery, probably
double-transgenic mice. Surprisingly, the resulting because of redundancy of the APP-like proteins.
offspring suffered from a very high premature mor- However, it is believed that the secreted form of
tality rate, with lifespans of between 6 and 20 wk, APP plays important roles in neuronal plasticity
preventing an extensive analysis of the phenotype and survival (Mattson, 1997).
of adult, and especially aged mice (unpublished Most recently, this role became even more puz-
results). In the absence of any major organic defects, zling, since APP was implemented as a kinesin-1
we have now confirmed that the APP x tau double- receptor (Kamal et al., 2000; 2001). APP apparently
transgenic mice appear to be extremely sensitive to binds to the light chain of kinesin-1, which itself is
external environmental stress, not unlike some responsible for anterograde axonal transport and
patients. A possible related vulnerability to stress consists of two light chains (KLC) associated with
was previously reported in a study of one line of two heavy chains (KIF5B). KIF5B contains ATP- as
APP-mutant-transgenic mice (Pedersen et al., 1999). well as microtubule-binding motifs that are essen-
Analysis of young double-transgenic mice did not tial for vesicle transport. KLC associates with KIF5B
demonstrate a major synergistic effect of pathology, and tethers membrane vesicles containing a subset
which was also largely lacking in the parent strains of proteins. Kinesin takes care of anterograde axonal
at young ages (less than 3 mo, unpublished results). transport and dynein is responsible for retrograde
Others have demonstrated an interaction axonal transport, but the structure of dynein is not
between APP and tau protein in double-transgenic examined here.
mice, using a mutant form of tau protein, as opposed An axonal membrane compartment was isolated
to the wild-type human tau that we have used (Lewis from sciatic nerve and corpus callosum by gradient
et al., 2001). These mice develop signs of additional centrifugation and binding to an antibody against
if not synergistic pathology, whereby NFTs APP that contained BACE and presenelin-1 (PS1), as
become apparent in regions of the brain that are not well as the degradation products of APP, Aβ40, and
affected in single-tau-mutant mice. The effect Aβ42 (Kamal et al., 2001). Isolated fragments of sci-
appeared to be mediated through projection of atic nerve, when incubated at 37°C, were shown to
axonal extensions, which was interpreted to mean generate Aβ40 and Aβ42 and increased soluble
that Aβ-mediated neurotoxicity by axonal projec- kinesin-1 and particulate Aβ40 and Aβ42 were found
tion, implicating axonal transport in increasing vul- in sciatic nerve segments after ligation. The combined
nerability of neurons to Aβ in a “feed-forward” data are taken as strong evidence that processing of
fashion. How this would result in increased tangle APP to Aβ can occur in axonal vesicles transported
formation is open for debate and more experimen- by kinesin-1 (Kamal et al, 2001). This would consti-
tation. It is in agreement with the proposal that tute the first link of axonal transport involving APP
plaque and tangle formation can begin in different and Aβ generation on the one hand and the micro-
brain areas, and the presence of amyloid causes tubular molecular motors on the other, indirectly
spreading of tangle pathology along neuronal tracts implicating tau protein.
in a hierarchal fashion—or vice versa (Braak and In a completely different model, deletion of the
Braak, 1991; Delacourte et al., 1999). APP-like gene (Appl) or overexpression of human
APP or APPL in Drosophila caused axonal-
transport phenotypes similar to kinesin and dynein
A Role for APP in Axonal Transport— mutants (Gunawardena and Goldstein, 2001). Dele-
Or for Axonal Transport tion of APPL resulted in vesicle or organelle stalling
in axons of Drosophila larva. Overexpression of
in APP Metabolism? APP695 or APPL led to non-APP-vesicle stalling
and axonal accumulations. Deletion of the
The biological function of APP remains an C-terminus of APP695 or APPL, including the
enigma, since it was identified as the precursor of kinesin-binding region, disrupted axonal transport
the Aβs and already then shown to be a proteinase of APP695 and APPL.
inhibitor, at least in regard to the isoforms that con- Although these findings are most interesting and
tain the Kunitz inhibitor domain. Without entering above all intriguing, they do not yet offer a solution

NeuroMolecular Medicine Volume 2, 2002


Axonal Transport in Alzheimer’s Disease 159

to the impasse we entered: the physiological role and production of APP in Down’s syndrome is in
of mammalian APP. On the contrary. If APP medi- perfect agreement with the amyloid-β cascade
ates its own transport and that of its complete hypothesis, which demands merely increased Aβ
Aβ-metabolizing machinery—e.g., BACE and PS1— production but not necessarily disturbed axonal
in vesicles along the axons, the question is, for what transport. Other systems and arguments have already
purpose? One possibility is that the destination of been addressed. Direct interaction between APP and
the transported vesicles is determined by proteol- tau in the same neuron does not appear to be a pre-
ysis of APP, which only complicates matters further requisite for the combined lesions to occur. There-
because local signals along the axon will have to be fore, the synergism between tangle and amyloid
invoked. pathology may well be regarded as “distant,” as sup-
ported by some experimental evidence (Lewis et al.,
2001; Götz et al., 2001) as well as by neuropatholog-
Is an Axonal Transport Deficit ical observations in AD (Braak and Braak, 1994).
Inherently Involved in AD Pathology?
Axonal transport cannot be investigated directly A Concluding, Not Conclusive
in humans, but neuropathological observations of Scheme
AD patients have been interpreted to suggest a prob-
lem. Electron microscopy showed a disturbed neu- A convincing scenario could consist of an initial
ronal cytoskeleton in AD brain (Perry et al., 1991; separate formation of Aβ peptides and hyper-
and references therein), which evidently could be phosphorylation of tau, eventually resulting in
consequent to the pathology, not its cause or medi- plaques and tangles respectively, but essentially
ator. The very few studies that used biopsy mater- by a “spreading” phenomenon via axonotoxicity
ial provided shreds of evidence for a reduction in of Aβ peptides in any form in which they can
the number of microtubules in affected neurons, exist, Aβ-(proto)fibrils, Aβ-oligomers, or ADDLs
circumventing at least partially the postmortem (Lambert et al., 1998; Koo et al., 1999; Walsh et al.,
problem of dissolution of microtubules. 2000), synergizing both aspects of the pathology.
In the absence of direct evidence in patients, the Inherently, the increased Aβ toxicity somehow leads
model systems have offered possibilities as to how to increased tangle formation—e.g., by hyperphos-
axonal transport might become impaired in AD: (i) phorylation of tau. This further disrupts axonal
by increased APP production, (ii) by increased tau transport and results in increased neuronal vul-
protein production, (iii) by increased protein tau nerability, and evidently, this is a vicious circle (see
phosphorylation, or (iv) by neuronal expression of Figs. 3A, 4).
apolipoprotein (ApoE) (see Fig. 3). The first two pos- Some evidence for this scheme has been found
sibilities have not been substantiated unequivocally in hippocampal cell cultures derived from tau
in the AD brain in later stages, let alone identified protein-knockout and tau-transgenic mice, demon-
as the primary event. Thus, it must be considered strating that tau was needed for Aβ toxicity in
that normal expression of tau and APP in combi- hippocampal cultures (Rapoport et al., 2002). Phar-
nation with age-related changes is sufficient to cause macological inhibition of MAPK blocked neurite
disturbances in axonal transport in AD, shifting the retraction of neurons exposed to Aβ (Rapoport and
problem again to be defined as “age-related Ferreira, 2000), and brain slices from Fyn-knockout
changes” in molecular terms! mice were completely protected from ADDL toxic-
Still, other arguments that have been put forth as ity (Lambert et al., 1998)—an interesting link, since
to why the transport function of APP could be related MAPK is positioned downstream from tyrosine
to the initiation of AD pathology are: (i) apposition kinase Fyn (Williamson et al., 2002). Another inter-
of APP, BACE, and PS in the same vesicle, (ii) extra esting link is offered by Niemann-Pick disease, since
APP gene copy in Down syndrome, (iii) dystrophic these patients have tangles that are indistinguish-
neurites in Drosophila that overexpress APP (Gold- able from the NFTs in AD. The product of the
stein et al., 2001). Although these arguments point responsible Niemann-Pick gene (NPC) regulates
to and offer possibilities, none separately or in com- cholesterol homeostasis, and transgenic mice over-
bination are totally convincing. The extra gene copy expressing NPC1 show cognitive defects, motoric

NeuroMolecular Medicine Volume 2, 2002


160 Terwel et al.

Fig. 3. Schematic representation of axonal transport of vesicles, incorporating the major molecular players
involved in AD. (A) A vesicle containing APP, BACE, and PS1 is transported along the axonal microtubules, even-
tually delivering Aβ peptides and/or aggregates to the synapse (based on Kamal et al., 2001). The resulting neu-
rotoxic signals are mediated via different pathways into nearby and distant neurons to which they project, and
trigger hyperphosphorylation of tau by specified kinases (see text for details). The depletion of active tau protein
destabilizes the microtubular network and further disturbs axonal transport. (B) In normal conditions (upper)
diverse organelles (red circles) and APP-carrying vesicles (open circles) are axonally transported and arrive at the
synapse. Increasing the levels of tau protein inhibits kinesin motors to bind to the microtubules, and less vesicles
of all types arrive at the synapse: axonal transport is impaired (middle). When the levels of APP are increased, it
is anticipated (Kamal et al., 2001) that vesicles carrying APP would preferentially bind to kinesin motors, and
thereby less organelles and more vesicles carrying APP, with higher loads of Aβ, will arrive at the synapse (lower).
Color image available for viewing at www.humanapress.com

NeuroMolecular Medicine Volume 2, 2002


Axonal Transport in Alzheimer’s Disease 161

Fig. 4. Scheme relating unknown causes of sporadic AD to the pathological lesions and impaired axonal trans-
port. The unknown causes of sporadic AD, represented by X and Y, affect separately or combined the processing
of APP and the control of phosphorylation of tau protein, leading respectively to increased production of amyloid-
fragments of APP (e.g., C99, Aβ40, and Aβ42) and to hyperphosphorylation of tau protein (tau-P). These events
give rise in the first (or in last) instance to the well-known amyloid and tau-pathology of plaques and tangles that
are the postmortem hallmarks of all AD patients. The link of hyperphosphorylation of tau to microtubular desta-
bilization and impaired axonal transport can be taken established, as opposed to the inference of C99 and/or Aβ
peptides with tau-phosphorylation, or vice versa (indicated by the double-headed arrow with question mark). The
theory that impaired axonal transport negatively contributes to the pathology by augmenting the generation of Aβs
has not yet been proven, as indicated by the question mark. Other question marks refer to problems and questions
raised and expanded in the text. Color image available for viewing at www.humanapress.com

problems with hyperphosphorylation of tau, and tangle formation involves GSK-3β. The mouse
a 10-fold increase in MAPK activity (Sawamura et models do not unequivocally answer the question
al., 2001; Voikar et al., 2002). It appears unlikely (Lucas et al., 2001; Spittaels et al., 2000). In the brain
that the known cholesterol export problem in these of AD patients, active GSK-3β is associated with
patients relates to the problem caused by the ApoE4 the earliest signs of tangle formation (Pei et al.,
allele in AD patients. On the other hand, the direct 1999) and in a paradigm of neuronal apoptosis
test of the hypothesis of neuronal expression of in vivo—e.g., hypometabolism induced by a
ApoE4 in transgenic mice resulted in very similar glutamate antagonist, GSK-3β activation preceded
motoric problems as in tau-4R transgenic mice, tau hyperphosphorylation (Elyaman et al., 2002).
including the progressive accumulation of hyper- Most recently, co-overexpression of tau and shaggy,
phosphorylated tau with age (Tesseur et al., the GSK-3β homolog in Drosophila, resulted in
2000a,b). This unexpected finding must be further neurofibrillary pathology in transgenic flies
analyzed to define the signaling pathways involved, (Jackson et al., 2002). The circumstances that
especially with regards to the ApoER2/LRP8 increase or deregulate GSK-3β activity in AD are
receptor as essential reelin receptor during unclear, but are likely related to cellular stress asso-
development. ciated mainly with aging as the major risk factor
Further experimental evidence is needed to for AD. This brings us back to defining the “aging”
determine whether the signaling pathways and problem in molecular terms.
kinases discussed here are indeed as prominently In conclusion, although it is most attractive
involved as we have proposed. Nevertheless, a to postulate that the pathology in AD is from the
major problem remains—not if, but how early onset accompanied by disturbed axonal transport,

NeuroMolecular Medicine Volume 2, 2002


162 Terwel et al.

its acceptance as a primary event appears dif- Braak E. and Braak E. (1991) Neuropathological stage-
ficult to defend. On the other hand, the secondary ing of Alzheimer-related changes. Acta Neuropathol.
effects on metabolism and synaptic function make 82, 239–259.
it a likely partner in the subsequent stages of the Braak E., Braak H., and Mandelkow E.-M. (1994) A
pathology of AD and other neuro-degenerative dis- sequence of cytoskeletal changes related to the for-
orders. Whereas a stronger case can be made now mation of neurofibrillary tangles and neuropil
than before for linking Aβ toxicity with tau phos- threads. Acta Neuropathol. 87, 554–567.
phorylation and axonal transport disturbances, Buée L., Bussière T., Buée-Scherrer V., Delacourte A.,
future research should be directed at understand- and Hof P. R. (2000) Tau protein isoforms, phos-
ing the mechanisms involved, to eventually allow phorylation and role in neurogenerative disorders.
Brain Res. Rev. 33, 95–130.
some remediation.
Caceras A. and Kosik K. S. (1990) Inhibition of neurite
polarity by tau antisense oligonucleotides in pri-
Acknowledgments mary cerebellar neurons. Nature 343, 461–463.
Caceras A., Potrebic S., and Kosik K. S. (1991) The effect
of tau antisense oligonucleotides on neurite for-
This investigation was supported by Fonds voor mation of cultured cerebellar macroneurons.
Wetenschappelijk Onderzoek-Vlaanderen (FWO- J. Neurosci. 11, 1515–1523.
Vlaanderen); the EEC-5th Framework Program; Delacourte A. and Buée L. (2000) Tau pathology: a
the Rooms-Fund; the Janssen Research Foundation; marker of neurodegenerative disorders. Curr. Opin.
the KULeuven GOA-Research Fund and KULeu- Neurol. 13, 371–376.
ven R&D; the Institute voor Aanmoediging van Delacourte A., David J. P., Sergeant N., et al. (1999) The
Wetenschappelijk en Technisch Onderzoek (IWT). biochemical pathway of neurofibrillary degenera-
Ilse Dewachter is a post-doctoral fellow of FWO- tion in aging and Alzheimer’s disease. Neurology
Vlaanderen and Dick Terwel is a post-doctoral 52, 1158–1165.
fellow of IWT. Dewachter I., Van Dorpe J., Smeijers L., Gilis M., Kuiperi
C., Laenen I., et al. (2000) Aging increased amyloid
peptide and caused amyloid plaques in brain of
References old APP/V717I transgenic mice by a different
mechanism than mutant presenilin 1. J. Neurosci. 20,
Alonso A. C., Grundke-Iqbal I., and Iqbal K. (1996) 6452–6458.
Alzheimer ’s disease hyperphosphorylated tau Dewachter I., Reverse D., Caluwaerts N., Ris L., Kuipéri
sequesters normal tau into tangles of filaments and C., Van den Haute C., et al. (2002) Neuronal
disassembles microtubules. Nat. Med. 2, 783–787. deficiency of presenilin 1 inhibits amyloid plaque
Alonso A. C., Zaidi T., Novak M., Grundke-Iqbal I., formation and corrects hippocampal long-
and Iqbal K. (2001) Hyperphosphorylation induces term potentiation but not a cognitive defect of
self-assembly of tau into tangles of paired helical amyloid precursor protein [V717I] transgenic mice.
filaments/straight filaments. PNAS 98, 6923–6928. J. Neurosci. 22, 3445–3453.
Baas P. W., Pienkowski T. P., and Kosik K. S. (1991) Drewes G., Lichtenberg-Kraag B., Doring F., Man-
Processes induced by tau expression in Sf9 cells delkow E.-M., Biernat J., Goris J., et al. (1992) Mito-
have an axon-like microtubule organization. J. Cell gen activated protein (MAP) kinase transforms
Biol. 115, 1333–1344. protein tau into an Alzheimer-like state. EMBO J.
Baumann K., Mandelkow E.-M., Biernat J., Piwnica- 11, 2131–2138.
Worms H., and Mandelkow E. (1993) Abnormal Drewes G., Mandelkow E.-M., Baumann K., Goris J.,
Alzheimer-like phosphorylation of protein tau Merlevede W., and Mandelkow E. (1993) Dephos-
by cyclin-dependent kinases cdk2 and cdk5. FEBS phorylation of protein tau and Alzheimer
Lett. 336, 417–424. paired helical filaments by calcineurin and
Biernat J. and Mandelkow E.-M. (1999) The develop- phosphatase-2A. FEBS Lett. 336, 424–432.
ment of cell processes induced by protein tau Drubin D. G. and Kirschner M. W. (1986) Protein tau
requires phosphorylation of serine 262 and 356 function in living cells. J. Cell Biol. 103, 2739–2746.
in the repeat domain and is inhibited by phospho- Ebneth A., Godeman R., Stamer K., Illenberger S.,
rylation in the proline-rich domains. Mol. Biol. Cell Trinczek B., Mandelkow E.-M., et al. (1998)
10, 727–740. Overexpression of protein tau inhibits kinesin-

NeuroMolecular Medicine Volume 2, 2002


Axonal Transport in Alzheimer’s Disease 163

dependent trafficking of vesicles, mitochondria, and Harada A., Oguchi K., Okabe S., Kuno J., Terada S.,
endoplasmatic reticulum: implications for Oshima T., et al. (1994) Altered microtubule orga-
Alzheimer’s disease. J. Cell Biol. 143, 777–794. nization in small-calibre axons of mice lacking tau
Esler W. P. and Wolfe M. S. (2001) A portrait of protein. Nature 369, 488–491.
Alzheimer secretases—new features and familiar Hasegawa M., Smith M. J., and Goedert M. (1998) Tau
faces. Science 293, 1449–1454. proteins with FTDP-17 mutations have reduced abil-
Elyaman W., Terro F., Wong N. S., and Hugon J. (2002) ity to promote microtubule assembly. FEBS Lett. 437,
In vivo activation and nuclear translocation of 207–210.
phosphorylated glycogen synthase kinase-3β in Hong M., Zhukareva V., Vogelsberg-Ragaglia V.,
neuronal apoptosis: links to tau phosphorylation Wszolek Z., Reed L., Miller B. I., et al. (1998)
Eur. J. Neurosci. 15, 651–660. Mutation-specific functional impairments in dis-
Friedhoff P., von Bergen M., Mandelkow E.-M., and tinct tau isoforms of hereditary FTDP-17. Science
Mandelkow E. (2000) Structure of protein tau and 282, 1914–1917.
assembly into paired helical filaments. BBA 1502, Hutton M., Lendon C. L., Rizzu P., Baker M., Froelich
122–132. S., Houlden H., et al. (1998) Association of
Geula C., Wu C. K., Saroff D., Lorenzo A., Yuan M., missense and 5′-splice-site mutations in tau with
and Yankner B. A. (1998) Aging renders the brain the inherited dementia FTDP-17. Nature 393,
vulnerable to amyloid-β protein neurotoxicity. Nat. 702–705.
Med. 4, 827–831. Iqbal K., Grundke-Iqbal I., Zaidi T., Merz P. A., Wen
Goedert M., Spillantini M. G., Potier M. C., Ulrich G. Y., Shaikh S. S., et al. (1986) Defective brain micro-
J., and Crowther R. A. (1989) Cloning and sequenc- tubule assembly in Alzheimer’s disease. Lancet 2,
ing of the cDNAencoding an isoform of microtubule- 421–426.
associated protein tau containing four tandem Ishihara T., Higuchi M., Zhang B., Yoshiyama Y., Hong
repeats: differential expression of protein tau M., Trojanowski J. Q., et al. (2001a) Attenuated neu-
mRNAs in human brain. EMBO J. 8, 393–399. rodegenerative disease phenotype in tau transgenic
Goedert M. and Jakes R. (1990) Expression of separate mouse lacking neurofilaments. J. Neurosci. 21,
isoforms of human tau protein: correlation with the 6026–6035.
tau pattern in brain and effects on tubulin poly- Ishihara T., Zhang B., Higuchi M., Yoshiyama Y.,
merisation. EMBO J. 9, 4225–4230. Trojanowski J. Q., and Lee V. M.-Y. (2001b) Age-
Goedert M., Hasegawa M., Jakes R., Lawler S., Cuenda dependent induction of congophilic neurofibrillary
A., and Cohen P. (1997) Phosphorylation of tau inclusions in tau transgenic mice. Am. J. Pathol.
microtubule-associated protein tau by stress- 158, 555–562.
activated protein kinases. FEBS Lett. 409, 57–62. Jackson G. R., Wiedau-Pazos M., Sang T.-K.,
Goldstein L. S. B. (2001) Kinesin molecular motors: Wagle N., Brown C. A., Massachi S., et al. (2002)
transport pathways, receptors, and human disease. Human wild-type tau interacts with wingless
PNAS 98, 6999–7003. pathway components and produces neuro-
Götz J., Chen F., Van Dorpe J., and Nitsch R. M. (2001) fibrillary pathology in Drosophila. Neuron 34,
Formation of neurofibrillary tangles in P301L tau 509–519.
transgenic mice induced by Aβ42 fibrils. Science 293, Johnson G. V. (1992) Differential phosphorylation of
1491–1495. tau by cyclic AMP-dependent protein kinase and
Greenwood J. A., Scott C. W., Spreen R. C., Caputo Ca2+/calmodulin-dependent protein kinase II: meta-
C. B., and Johnson G. V. (1994) Casein kinase II pref- bolic and functional consequences. J. Neurochem. 59,
erentially phosphorylates tau isoforms containing 2056–2062.
an amino-terminal insert. Identification of threo- Kamal A., Stokin G. B., Yang Z., Xia C.-H., and Gold-
nine 39 as the primary phosphate acceptor. J. Biol. stein S. B. (2000) Axonal transport of amyloid pre-
Chem. 269, 4373–4380. cursor protein is mediated by direct binding to the
Gunawardena S. and Goldstein L. S. B. (2001) Dis- kinesin light chain subunit of kinesin-1. Neuron 28,
ruption of axonal transport and neuronal viability 449–459.
by amyloid precursor protein mutations in Kamal A., Almenar-Queralt A., Leblanc J. F., Roberts
Drosophila. Neuron 32, 389–401. E. A., and Goldstein L. S. B. (2001) Kinesin-mediated
Hanemaaijer R. and Ginzburg I. (1991) Involvement axonal transport of a membrane compartment con-
of mature tau isoforms in the stabilization of neu- taining β-secretase and presenilin-1 requires APP.
rites in PC12 cells. J. Neurosci. 30, 163–171. Nature 414, 643–647.

NeuroMolecular Medicine Volume 2, 2002


164 Terwel et al.

Koo E. H., Lansbury P. T. Jr, and Kelly J. W. (1999) Amy- Michel G., Mercken M., Murayama M., Noguchi K.,
loid diseases: abnormal protein aggregation in neu- Ishiguro K., Imahori K., et al. (1998) Characteriza-
rodegeneration. PNAS 96, 9989–9990. tion of tau phosphorylation in glycogen synthase
Khatoon S., Grundke-Iqbal I., and Iqbal K. (1992) Brain kinase-3β and cyclin dependent kinase-5 activator
levels of microtubule-associated protein tau are ele- (p23) transfected cells. BBA 1380, 177–182.
vated in Alzheimer’s disease: a radioimmunoslot- Moechars D., Dewachter I., Lorent K., Reverse D.,
blot assay for nanograms of the protein. J. Neurochem. Baekelandt V., Naidu A., et al. (1999) Early pheno-
59, 750–753. typic changes in transgenic mice that overexpress
Ksiezak-Reding H., Binder L. I., and Yen S. H. (1988) different mutants of amyloid precursor protein in
Immunochemical and biochemical characterization brain. J. Biol. Chem. 274, 6483–6492.
of tau protein in normal and Alzheimer’s disease Morfini G., Szebenyi G., Elluru R., Ratner N., and Brady
brains with Alz 50 and Tau-1. J. Biol. Chem. 263, S. T. (2002) Glycogen synthase kinase 3 phospho-
7948–7953. rylates kinesin light chains and negatively regulates
Künzi V., Glatzel M., Nakano M. Y., Greber U. F., kinesin-based motility. EMBO J. 21, 281–293.
Van Leuven F., and Aguzzi A. (2002) Unhampered Nakazato Y., Sasaki A., Hirato J., and Ishida Y. (1984)
prion neuroinvasion despite impaired fast axonal Immunohistochemical localization of neurofila-
transport in transgenic mice overexpressing four- ment protein in neuronal degenerations. Acta
repeat tau. J. Neurosci. 22, 7471–7477. Neuropathol. (Berl.) 64, 30–36.
Lambert M. P., Barlow A. K., Chromy B. A., Edwards Norlund M. A., Lee J. M., Zainelli G. M., and Muma
C., Freed R., Liosatos M., et al. (1998) Diffusible, N. A. (1999) Elevated transglutaminase-induced
nonfibrillar ligands derived from Aβ1-42 are potent bonds in PHF tau in Alzheimer’s disease. Brain Res.
central nervous system neurotoxins. PNAS 95, 851, 154–163.
6448–6453. Nuydens R., Van den Kieboom G., Nolten C., Verhulst
Lee G., Neve R. L., and Kosik K. S. (1989) The micro- C., Van Osta P., Spittaels K., et al. (2002) Coexpres-
tubule binding domain of tau protein. Neuron 2, sion of GSK-3β corrects phenotypic aberrations of
1615–1624. dorsal root ganglion cells, cultured from adult
Lewis J., Dickson D. W., Lin W.-L., Chisholm L., Corral transgenic mice overexpressing human protein tau.
A., Jones G., Yen S.-H., et al. (2001) Enhanced Neurobiol. Dis. 9, 38–48.
neurofibrillary degeneration in transgenic mice Patrick G. N., Zukerberg L., Nikolic M., De La Monte
expressing mutant tau and APP. Science 293, S., Dikkes P., and Tsai L. H. (1999) Conversion of
1487–1491. p35 to p25 deregulates cdk5 activity and promotes
Lucas J. J., Hernandez F., Gomez-Ramos P., Moran neurodegeneration. Nature 402, 615–622.
M. A., Hen R. and Avila J. (2001) Decreased nuclear Pedersen W. A., Culmsee C., Ziegler D., Herman J. P.,
β-catenin, tau hyperphosphorylation and neu- and Mattson M. P. (1999) Aberrant stress response
rodegeneration in GSK-3β conditional transgenic associated with severe hypoglycemia in a trans-
mice. EMBO J. 20, 27–39. genic mouse model of Alzheimer’s disease. J. Mol.
Mandelkow E.-M., Biernat J., Drewes G., Gustke N., Neurosci. 13, 159–165.
Trinczek B., and Mandelkow E. (1995) Tau domains, Pei J. J., Braak E., Braak H., Grundke-Iqbal I., Iqbal K.,
phosphorylation, and interaction with micro- Winblad B., et al. (1999) Distribution of active glyco-
tubules. Neurobiol. Aging 16, 355–363. gen synthase kinase-3β (GSK-3β) in brains staged
Matsuo E. S., Shin R.-W., Billingsley M. L., Van de Voorde for Alzheimer disease neurofibrillary changes.
A., O´Connor M., Trojanowski J. Q., et al. (1994) J. Neuropathol. Exp. Neurol. 58, 1010–1019.
Biopsy-derived adult human tau is phosphorylated Perry G., Kawai M., Tabaton M., Onorato M., Mulvihill
at many of the same sites as Alzheimer’s disease P., Richey P., et al. (1991) Neuropil threads
paired helical filament tau. Neuron 13, 989–1002. of Alzheimer’s disease show a marked alteration of
Mattson M. P. (1997) Cellular actions of beta-amyloid the normal cytoskeleton. J. Neurosci. 11, 1748–1755.
precursor protein and its soluble and fibrillogenic Rapoport M. and Ferreira A. (2000) PD98059 pre-
derivatives. Physiol. Rev. 77, 1081–1132. vents neurite degeneration induced by fibrillar
Mattson M. P., Fu W., Waeg G., and Uchida K. (1997) β-amyloid in mature hippocampal neurons.
4-Hydroxynonenal, a product of lipid peroxida- J. Neurochem. 74, 125–133.
tion, inhibits dephosphorylation of the micro- Rapoport M., Dawson H. N., Binder L. I., Vitek M. P.,
tubule-associated protein tau. Neuroreport 8, and Ferreira A. (2002) Tau is essential to β-amyloid-
2275–2281. induced neurotoxicity. PNAS 99, 6364–6369.

NeuroMolecular Medicine Volume 2, 2002


Axonal Transport in Alzheimer’s Disease 165

Rouleau G. A., Clarke A. W., Rooke K., Pramatarova Stamer K., Vogel R., Thies E., Mandelkow E., and
A., Krizus A., Suchowersky O., et al. (1996) SOD1 Mandelkow E.-M. (2002) Tau blocks traffic of
mutation is associated with accumulations of neu- organelles, neurofilaments, and APP vesicles in
rofilaments in amyotrophic lateral sclerosis. Ann. neurons and enhances oxidative stress. J. Cell Biol.
Neurol. 39, 128–131. 156, 1051–1063.
Ruben G. C., Ciardelli T. L., Grundke-Iqbal I., and Iqbal Tesseur I., Van Dorpe J., Spittaels K., Van den Haute
K. (1997) Alzheimer disease hyperphosphorylated C., Moechars D., and Van Leuven F. (2000) Expres-
tau aggregates hydrophobically. Synapse 27, sion of human apolipoprotein E4 in neurons causes
208–229. hyperphosphorylation of protein tau in the brains
Sawamura N., Gong J.-S., Garver W. S., Heidenreich of transgenic mice. Am. J. Pathol. 156, 951–964.
R. A., Ninomiya H., Ohno K., et al. (2001) Site- Tesseur I., Van Dorpe J., Bruynseels K., Bronfman, F.,
specific phosphorylation of tau accompanied by Sciot R., Van Lommel A., et al. (2000) Prominent
activation of mitogen-activated protein kinase axonopathy and disruption of axonal transport in
(MAPK) in brains of Niemann-Pick type C mice. transgenic mice expressing human apolipoprotein
J. Biol. Chem. 276, 10,314–10,319. E4 in neurons of brain and spinal cord. Am. J. Pathol.
Scott C. W., Blowers D. P., Barth P. T., Lo M. M., Salama 157, 1495–1510.
A. I., and Caputo C. B. (1991) Differences in the abil- Trinczek B., Ebneth A., Mandelkow E.-M., and Man-
ities of human tau isoforms to promote microtubule delkow E. (1999) Tau regulates the attachment/
assembly. J. Neurosci. Res. 30, 154–162. detachment but not the speed of motors in
Shankar S. K., Yanagihara R, Garruto R. M., Grundke- microtubule-dependent transport of single
Iqbal I, Kosik K. S., and Gajdusek D. C. (1989) vesicles and organelles. J. Cell Sci. 112, 2355–2367.
Immunocytochemical characterization of neu- Van Dorpe J., Smeijers L., Dewachter I., Nuyens D., Spit-
rofibrillary tangles in amyotrophic lateral sclerosis taels K., Van den Haute C., et al. (2002) Promiment
and parkinsonism-dementia of Guam. Ann. Neurol. cerebral amyloid angiopathy in transgenic mice
25, 146–151. overexpressing the London mutant of human APP
Sisodia S. S. and St. George-Hyslop P. H. (2002) in neurons. Am. J. Pathol. 157, 1283–1298.
γ-secretase, Notch, Aβ and Alzheimer’s disease: Voikar V, Rauvala H, and Ikonen E. (2002) Cognitive
where do the presenilins fit in? Nature Rev. Neurosci. deficit and development of motor impairment in a
3, 281–290. mouse model of Niemann-Pick type C disease.
Spillantini M. G., Murrell J. R., Goedert M., Farlow Behav. Brain Res. 132, 1–10.
M. R., Klug A., and Ghetti B. (1998) Mutation in the Walsh D. M., Tseng B. P., Rydel R. E., Podlisney
tau gene in familial multiple system tauopathy with M. B., and Selkoe D. J. (2000) The oligomerization
presenile dementia. PNAS 95, 7737–7741. of amyloid β-protein begins intracellularly in cells
Spittaels K., Van den Haute C., Van Dorpe J., Bruynseels derived from human brain. Biochemistry 39,
K., et al. (1999) Prominent axonopathy in the brain 10,831–10,839.
and spinal cord of transgenic mice overexpressing Wang J. Z., Grundke-Iqbal I., and Iqbal K. (1996) Gly-
four-repeat human tau protein. Am. J. Pathol. 155, cosylation of microtubule-associated protein tau:
2153–2165. an abnormal posttranslational modification in
Spittaels K., Van den Haute C., Van Dorpe J., Geerts Alzheimer’s disease. Nat. Med. 2, 871–875.
H., Mercken M., Bruynseels K., et al. (2000) Glyco- Williamson R., Scales T., Clark B. R., Gibb G., Reynolds
gen synthase kinase-3β phosphorylates tau and res- C. H., Kellie S., et al. (2002) Rapid tyrosine phos-
cues axonopathy in the central nervous system of phorylation of neuronal proteins including tau and
human four-repeat tau transgenic mice. J. Biol. Chem. focal adhesion kinase in response to amyloid-β pep-
275, 41,340–41,349. tide exposure: involvement of src family protein
Spittaels K., Van den Haute C., Van Dorpe J., Terwel kinases. J. Neurosci. 22, 10–20.
D., Vandezande K., Lasrado R., et al. (2002) Neona- Yoshida H. and Ihara Y. (1993) Tau in paired helical fil-
tal neuronal over-expression of glycogen synthase aments is functionally distinct from fetal tau: assem-
kinase-3β reduces brain-size in transgenic mice. 113, bly incompetence of paired helical filament tau.
797–808. J. Neurochem. 61, 1183–1186.

NeuroMolecular Medicine Volume 2, 2002

You might also like