Tải bản đầy đủ (.pdf) (11 trang)

Tài liệu Báo cáo khoa học:Tyrosine phosphorylation of tau regulates its interactions with Fyn SH2 domains, but not SH3 domains, altering the cellular localization of tau ppt

Bạn đang xem bản rút gọn của tài liệu. Xem và tải ngay bản đầy đủ của tài liệu tại đây (224.3 KB, 11 trang )

Tyrosine phosphorylation of tau regulates its interactions
with Fyn SH2 domains, but not SH3 domains, altering the
cellular localization of tau
Alessia Usardi
1
, Amy M. Pooler
1
, Anjan Seereeram
1
, C. Hugh Reynolds
1
, Pascal Derkinderen
1
,
Brian Anderton
1
, Diane P. Hanger
1
, Wendy Noble
1,
* and Ritchie Williamson
1,2,
*
1 Department of Neuroscience, MRC Centre for Neurodegeneration Research, Institute of Psychiatry, King’s College London, UK
2 Biomedical Research Institute, Ninewells Medical School, University of Dundee, UK
Keywords
Fyn-SH2; Fyn-SH3; phosphorylation; tau;
tyrosine
Correspondence
R. Williamson, Biomedical Research
Institute, Ninewells Medical School,


University of Dundee, Dundee DD1 9SY, UK
Fax: +44 1382 740 359
Tel: +44 1382 740 347
E-mail:
*These authors contributed equally to this
work
(Received 19 April 2011, revised 20 May
2011, accepted 16 June 2011)
doi:10.1111/j.1742-4658.2011.08218.x
Recent reports have demonstrated that interactions between the microtu-
bule-associated protein tau and the nonreceptor tyrosine kinase Fyn play a
critical role in mediating synaptic toxicity and neuronal loss in response to
b-amyloid (Ab) in models of Alzheimer’s disease. Disruption of interactions
between Fyn and tau may thus have the potential to protect neurons from
Ab-induced neurotoxicity. Here, we investigated tau and Fyn interactions
and the potential implications for positioning of these proteins in membrane
microdomains. Tau is known to bind to Fyn via its Src-homology (SH)3
domain, an association regulated by phosphorylation of PXXP motifs in tau.
Here, we show that Pro216 within the PXXP(213–216) motif in tau plays an
important role in mediating the interaction of tau with Fyn-SH3. We also
show that tau interacts with the SH2 domain of Fyn, and that this associa-
tion, unlike that of Fyn-SH3, is influenced by Fyn-mediated tyrosine phos-
phorylation of tau. In particular, phosphorylation of tau at Tyr18, a reported
target of Fyn, is important for mediating Fyn-SH2–tau interactions. Finally,
we show that tyrosine phosphorylation influences the localization of tau to
detergent-resistant membrane microdomains in primary cortical neurons,
and that this trafficking is Fyn-dependent. These findings may have implica-
tions for the development of novel therapeutic strategies aimed at disrupting
the tau ⁄ Fyn-mediated synaptic dysfunction that occurs in response to ele-
vated Ab levels in neurodegenerative disease.

Structured digital abstract
l
Fyn physically interacts with tau by pull down (View interaction)
l
Fyn physically interacts with tau by pull down (View interaction)
Introduction
The microtubule-associated protein tau is a predomi-
nantly neuronal soluble phosphoprotein that is mainly
cytoplasmic, but is also present in nuclear [1,2]
and membrane [3–5] compartments of various cell
types. Abnormalities in tau, including its aberrant
phosphorylation, truncation and aggregation, are
causally associated with neuronal loss in a family of
neurodegenerative disorders named the tauopathies,
which include Alzheimer’s disease (AD), progressive
supranuclear palsy and frontotemporal dementia with
Abbreviations
AD, Alzheimer’s disease; Ab, b-amyloid; CHO, Chinese hamster ovary; CNS, central nervous system; DRM, detergent-resistant
microdomain; GST, glutathione-S-transferase; NMDA, N-methyl-
D-aspartate; PSD, postsynaptic density; SEM, standard error of the mean;
SH, Src homology.
FEBS Journal 278 (2011) 2927–2937 ª 2011 The Authors Journal compilation ª 2011 FEBS 2927
Parkinsonism associated with tau mutations on chro-
mosome 17 [6,7]. In AD, tau is believed to act in syn-
ergy with b-amyloid (Ab) to mediate neuronal loss [8].
Recently, we and others have highlighted the impor-
tance of tau interactions with the membrane-anchored
nonreceptor tyrosine kinase Fyn during A b-mediated
neurodegeneration in cell and animal models of AD
[9–12].

Ab-induced neurotoxicity in primary cultured neu-
rons is dependent upon both Fyn and tau [9,13]. This
toxicity is associated with the accumulation of Ab on
plasma membranes and the recruitment of tau into
lipid rafts [9], where tau is phosphorylated on the
putative Fyn residue Tyr 18 [14]. In addition, interac-
tions between Fyn and tau are important for the den-
dritic positioning of these proteins, a localization that
has been shown to be a critical factor for Ab-induced
toxicity [11]. In a transgenic mouse model of AD over-
expressing mutant human amyloid precursor protein,
tau-dependent positioning of Fyn in dendrites appears
to regulate Fyn activation in response to Ab [11].
Subsequent Fyn-dependent stabilization of N-methyl-
D-aspartate (NMDA) receptor interaction with the
postsynaptic density (PSD) protein PSD-95 results in
Ab-induced excitotoxicity [11]. Furthermore, Fyn
sensitizes mice to the toxic effects of Ab [15,16], and
tau is required for the Fyn-mediated and Ab-mediated
synaptic deficits and network impairments observed in
other mouse models of AD [12], further supporting the
idea that disruption of Fyn–tau interactions may have
therapeutic utility in AD.
Fyn can phosphorylate tau directly on Tyr18, one of
five tyrosines present on tau [17–19], and tau also
interacts with Fyn via its Src homology (SH)3 domain
[20,21]. The SH3 domain of Fyn binds to proline-rich
motifs within the sequence PXXP in interacting pro-
teins. Seven such motifs are present in the proline-rich
domain of the longest isoform of tau in the human

central nervous system (CNS). Two distinct PXXP
motifs in tau, residing within residues 213–219 and
233–236, have been suggested to mediate its associa-
tion with Fyn-SH3 [20,21], and this interaction is regu-
lated by the serine ⁄ threonine phosphorylation status of
tau [21].
Fyn also interacts with proteins through its SH2
domain, which recognizes phosphorylated tyrosines on
target proteins [22,23]. Such interactions regulate the
induction of several signal transduction pathways [24]
that could play a role in Ab-induced neuronal loss.
Tau is known to be tyrosine phosphorylated in post-
mortem AD brain [17,18,25] as well as in transgenic
mouse models of tauopathy, in which tyrosine phos-
phorylated tau is associated with the development of
tau pathology and neuronal loss [26]. Thus, it is
important to establish whether or not tau interacts
with Fyn-SH2, as this would probably reveal that the
tyrosine phosphorylation status of tau is important for
mediating the association of these two proteins.
Here, we used Chinese hamster ovary (CHO) cells to
characterize the interaction between exogenously
expressed human wild-type tau and Fyn. Previous
studies using truncated tau constructs have indicated
that Fyn-SH3 interactions are mediated by either
Pro216 or Pro233 on tau [20,21]. Using mutant forms
of full-length tau, we show that direct interaction with
Fyn-SH3 is mediated predominantly by Pro216 in tau.
In addition, we show that tau interacts with Fyn-SH2
and that tyrosine phosphorylation of tau, particularly

on Tyr18, mediates this binding. Furthermore, we
show that Fyn-mediated tyrosine phosphorylation of
tau is important for its recruitment to detergent-resis-
tant microdomains (DRMs) on primary neurons.
These findings suggest that the tyrosine phosphoryla-
tion-dependent interactions of tau and Fyn may play
an important role in regulating the cellular localization
of Fyn and tau.
Results
Tau interacts with Fyn-SH2
Because tau contains tyrosines that could be targeted
by Fyn, and tyrosine phosphorylation influences Fyn-
SH2 binding to target proteins, we set out to deter-
mine whether or not tau binds to Fyn-SH2.
CHO cells were transiently cotransfected with plas-
mids expressing V5-tagged human 2N4R tau, the lon-
gest isoform of tau present in the adult human CNS,
and Fyn. Following transfection, CHO cells were trea-
ted either with pervanadate, to prevent the dephos-
phorylation of tyrosines, or with catalase as a control.
Pervanadate is a cell-permeable inhibitor of protein
tyrosine phosphatases that acts by irreversible oxida-
tion of the catalytic site [27]. We have previously dem-
onstrated that pervanadate increases tyrosine
phosphorylation of several proteins, including tau, in
cell lines [25]. CHO cell lysates were then incubated
with glutathione-S-transferase (GST)–Fyn-SH2 and
GST–Fyn-SH3 fusion proteins linked to glutathione
Sepharose beads, and bound proteins were collected.
An antibody directed against total (phosphorylated

and nonphosphorylated) tau revealed a primary band
of  64 kDa, corresponding to V5-tagged tau, on
western blots of cell lysates (Fig. 1A). Tau was also
detected in the GST–Fyn-SH2-bound fraction, but not
in the GST-only-bound fraction pulldowns (Fig. 1A).
Tyrosine phosphorylation-dependent tau–Fyn binding A. Usardi et al.
2928 FEBS Journal 278 (2011) 2927–2937 ª 2011 The Authors Journal compilation ª 2011 FEBS
This indicates that a proportion of tau interacts with
Fyn-SH2 and that this binding is specific, as it is not
related to the presence of GST. Western blotting with
a polyclonal antibody against GST confirmed that an
equal amount of GST–Fyn-SH2 beads was used in
each pulldown. Pervanadate treatment of CHO cells
coexpressing tau and Fyn resulted in an increased
amount of tau bound to Fyn-SH2, as compared with
cells treated with catalase (Fig. 1A). Furthermore, a
tau species of  68 kDa was apparent in SH2 pull-
downs from pervanadate-treated cells that had been
transfected with tau. This may represent a more highly
phosphorylated tau species or might indicate a differ-
ent conformation of tau with reduced electrophoretic
mobility. Densitometric analysis of GST–Fyn-SH2-
bound tau, as a proportion of total tau in each cell
lysate, revealed that pervanadate increased Fyn-SH2–
tau binding by approximately six-fold as compared
with controls (Fig. 1B). These results show that inhibi-
tion of tyrosine phosphatases with pervanadate results
in significantly increased binding of tau to Fyn-SH2,
thus suggesting that the interaction between tau and
Fyn-SH2 is enhanced by increased tyrosine phosphory-

lation in cells.
Tyr18 of tau plays an important role in the
interaction of tau with Fyn-SH2
To determine whether the tyrosine phosphorylation
status of tau itself is important for its interaction with
Fyn-SH2, CHO cells were transiently cotransfected
with wild-type tau or a mutant construct in which all
five tyrosines in tau (Tyr18, Tyr29, Tyr197, Tyr310
and Tyr394) were replaced with phenylalanine, gener-
ating YallF tau. This mutant tau species is therefore
unable to be phosphorylated on tyrosines. In addition,
to determine whether phosphorylation at individual
tyrosines in tau is important for its interaction with
Fyn-SH2, CHO cells were transiently cotransfected
with Fyn together with one of five mutant tau con-
structs in which single tyrosines were mutated to phen-
ylalanine (Y18F, Y29F, Y197F, Y310F or Y394F).
Transfected CHO cells were treated with pervanadate
or catalase, as above, prior to pulldown with GST–
Fyn-SH2 and determination of bound proteins on
western blots.
Immunolabelling with an antibody directed against
tau confirmed previous findings that pervanadate treat-
ment increased the association of wild-type tau with
GST–Fyn-SH2. In addition, although a 64-kDa YallF
tau band was apparent in lysates from cells cotrans-
fected with the Fyn construct, there were only trace
amounts of YallF tau bound to GST–Fyn-SH2
(Fig. 2A). Moreover, the association of YallF tau with
Fyn-SH2 was not influenced by pervanadate. These

results show that prevention of tau tyrosine phosphor-
ylation almost completely ablates the ability of tau to
bind to Fyn-SH2, indicating that this interaction is
dependent on tyrosine phosphorylation of tau. A small
proportion of expressed Y18F, Y29F, Y197F, Y310F
and Y394F tau each bound to Fyn-SH2 under control
conditions, and this binding was elevated with per-
vanadate (Fig. 2A). Notably, Y18F tau appeared to be
less able than Y29F, Y197F, Y310F and Y394F tau to
bind GST–Fyn-SH2, suggesting that phosphorylation
of tau at Tyr18 may be particularly important for its
interaction with Fyn-SH2 (Fig. 2B). Western blotting
with a polyclonal antibody against GST confirmed
that the same amount of GST–Fyn-SH2 beads was
used in each pulldown.
The amount of tau bound to GST–Fyn-SH2 follow-
ing pervanadate treatment was quantified as a propor-
tion of tau in cell lysates (Fig. 2B). These results
revealed that tau binding to Fyn-SH2 was almost com-
pletely ablated when all tyrosines on tau were substi-
tuted (YallF). In contrast, all of the tau mutants with
substitutions of individual tyrosines were able to bind
GST–Fyn-SH2 to some extent. Y18F tau showed
Fig. 1. Tyrosine phosphorylation of tau increases its association
with Fyn-SH2. CHO cells were cotransfected with plasmids
expressing Fyn and V5-tagged wild-type tau. Cells were treated
with 100 l
M pervanadate (P) or catalase (C). (A) CHO cell lysates
and proteins pulled down by GST or GST–Fyn-SH2 beads on wes-
tern blots labelled with antibodies against tau (V5) or GST. Num-

bers on the left indicate molecular masses (kDa). (B) Bar chart
showing the proportion of total tau bound to Fyn-SH2 in CHO cells
treated with pervanadate or catalase as mean ± SEM. N =4.
***P < 0.005.
A. Usardi et al. Tyrosine phosphorylation-dependent tau–Fyn binding
FEBS Journal 278 (2011) 2927–2937 ª 2011 The Authors Journal compilation ª 2011 FEBS 2929
significantly reduced association with GST–Fyn-SH2,
as compared with wild-type tau (P < 0.01), whereas
phenylalanine substitutions of Tyr29, Tyr197, Tyr310
or Tyr394 in tau did not significantly influence the
interaction with Fyn-SH2. These results suggest that
Tyr18, the putative Fyn kinase site on tau, plays a
major role in mediating interactions between tau and
Fyn-SH2. However, a contribution from other tyro-
sines cannot be excluded, as a greater proportion of
Y18F than of YallF mutant tau cosedimented with
GST–Fyn-SH2.
Interestingly, whereas pervanadate induced the
appearance of an  68-kDa band in wild-type, Y18F,
Y29F, Y197F and Y310F tau, this band was not
apparent on western blots of lysates of Y394F tau
(Fig. 2A). The absence of this larger species could indi-
cate that phosphorylation of tau by Fyn is reduced
and therefore, in addition to phosphorylation by c-Abl
[25], Fyn might also target Tyr394 in tau. Indeed, Fyn
has previously been reported to phosphorylate both
Tyr18 and Tyr394 [19], although it is clear that Tyr394
is phosphorylated predominantly by c-Abl. However,
impaired phosphorylation of Tyr394 did not appear
to influence the binding of tau to Fyn-SH2, as there

was no significant difference in the amount of Y394F
that cosedimented with GST–Fyn-SH2 when compared
with wild-type tau. This may be possible because,
although Fyn-SH2 binds directly to phosphotyro-
sines, the amino acid sequence context of the phosp-
hotyrosine site is also important in SH2 domain
recognition, a property that allows SH2 domains to
display binding preferences for specific sites on target
proteins [28].
Tyrosine phosphorylation does not modulate tau
binding to Fyn-SH3
We and others have previously demonstrated that Fyn
binds to tau predominantly through its SH3 domain,
and this interaction is regulated by serine ⁄ threonine
phosphorylation of tau [20,21]. To determine whether
the tyrosine phosphorylation status of tau also affects
the binding of tau to Fyn-SH3, CHO cells were
cotransfected with Fyn together with either wild-type
or the YF mutant forms of tau. Cell lysates containing
equal amounts of tau were subjected to pulldown
assays with GST–Fyn-SH3, and GST-bound proteins
were then assessed by immunoblotting.
Western blotting of lysates from pervanadate-treated
cells with an antibody against total tau revealed
decreased electrophoretic mobility of tau, with the
appearance of an  68-kDa tau species in wild-type and
all of the mutant forms of tau except for YallF and
Y394F tau (Fig. 3). In contrast to the results obtained
with Fyn-SH2, wild-type tau and all of the YF mutant
tau proteins were detected following pulldown with

GST–Fyn-SH3 beads. There were no significant differ-
ences in the proportion of wild-type or mutant YF tau
associated with Fyn-SH3. Furthermore, treatment of
Fig. 2. The phosphorylation status of Tyr18 on tau is important for
Fyn-SH2–tau interaction. CHO cells were cotransfected with Fyn
and V5-tagged wild-type (WT) or mutant YallF, Y18F, Y29F, Y197F,
Y310F or Y394F tau. Cells were treated with 100 l
M pervanadate
(P) or catalase (C). (A) CHO cell lysates and proteins pulled down
by GST–Fyn-SH2 beads on western blots labelled with antibodies
against tau (V5) or GST. Numbers on the left indicate molecular
masses (kDa). (B) Bar chart showing the proportion of total tau
bound to Fyn-SH2 in CHO cells expressing wild-type or mutant tau.
Mean values ± SEM are shown relative to the amount of wild-type
tau bound to Fyn-SH2. N =3.**P < 0.01, ***P < 0.005.
Fig. 3. Tau interactions with Fyn-SH3 are not influenced by tyro-
sine phosphorylation. CHO cells were transiently cotransfected
with Fyn and V5-tagged wild-type (WT) or mutant YallF, Y18F,
Y29F, Y197F, Y310F or Y394F tau. Cells were treated with pervana-
date (P) or catalase (C). Cell lysates and proteins pulled down by
GST–Fyn-SH3 beads were assessed on western blots labelled with
antibodies against V5 or GST. Numbers on the left indicate molecu-
lar masses (kDa). N =3.
Tyrosine phosphorylation-dependent tau–Fyn binding A. Usardi et al.
2930 FEBS Journal 278 (2011) 2927–2937 ª 2011 The Authors Journal compilation ª 2011 FEBS
CHO cells with pervanadate did not affect the interaction
of wild-type or mutant tau with Fyn-SH3 (Fig. 3). These
findings indicate that, unlike the case for Fyn-SH2, tyro-
sine phosphorylation does not play a role in mediating
interactions between tau and Fyn-SH3, and this further

suggests that different, and possibly independent, mech-
anisms are involved in these interactions of tau with the
same protein.
Interactions with Fyn-SH3 are regulated by key
PXXP motifs in tau
Tau has been shown to bind Fyn-SH3 through spe-
cific PXXP motifs, seven of which are present in
tau. Six of these occur as three pairs of partially over-
lapping tandem sequences (tau residues Pro176–
Pro182, Pro200–Pro206 and Pro213–Pro219), and the
seventh exists as a separate motif at Pro233–Pro236.
However, there is some discrepancy over which of
these PXXP motifs is most important for tau binding
to Fyn-SH3. In neuroblastoma cells, truncated tau
mutants lacking Pro233–Pro236 were used to demon-
strate that this region of tau is critical for the binding
of tau to Fyn-SH3 [20]. Conversely, using synthetic
peptides, we found that Fyn binds strongly to Pro213–
Pro219 of tau, but exhibits little interaction with
Pro233–Pro236 [21]. To further investigate which
PXXP motifs in tau are responsible for Fyn-SH3
binding, we generated alanine-substituted tau mutant
constructs, P216A and P233A, for V5-tagged wild-type
human tau.
CHO cells were cotransfected with Fyn together
with V5-tagged wild-type, P216A mutant or P233A
mutant tau. Cell lysates were analysed on western blots
probed with an antibody against V5 to confirm the
equivalence of tau protein expression in CHO cells
(Fig. 4A). GST–Fyn-SH3 beads were used to pull

down bound tau in cell lysates. Detection of bound
proteins by immunoblotting with an antibody against
V5 revealed tau bands of  64 kDa in the GST–Fyn-
SH3-bound fraction from cells transfected with each of
the tau constructs. Densitometric analysis of Fyn-SH3-
bound tau as a proportion of that in corresponding
cell lysates showed that significantly less P216A tau
was pulled down by GST–Fyn-SH3 than by either
wild-type or P233A tau (P < 0.05). There were no sig-
nificant differences between the amounts of wild-type
or P233A tau bound by the GST–Fyn-SH3 beads
(Fig. 4B). These results indicate that tau Pro216 plays
an important role in mediating the binding of full-
length wild-type tau to Fyn-SH3, in agreement with
the findings of our previous study with synthetic tau
peptides [21].
Tyrosine phosphorylation influences tau content
in neuronal DRMs
Interactions between tau and Fyn are important for
the cellular distribution of these proteins. For example,
we have shown that the trafficking of tau to DRMs [9]
and plasma membranes [5] is Fyn-dependent. As we
found that the tyrosine phosphorylation of tau influ-
ences its interaction with Fyn, we therefore set out to
investigate the role of tyrosine phosphorylation in
intracellular tau trafficking.
Primary cortical neurons cultured from wild-type
(Fig. 5A) and Fyn-deficient (Fig. 5B) mice, with
matched genetic backgrounds, were treated with per-
vanadate or catalase, as above. Cell homogenates were

collected, and DRMs were isolated and concentrated.
Western blotting with an antibody against the DRM
marker flotillin-1 was used to demonstrate that DRMs
were successfully isolated from wild-type and Fyn-defi-
cient mice (Fig. 5). Similarly, immunoblotting with an
antibody against Fyn revealed its enrichment in DRMs
prepared from wild-type neurons, but not from neu-
rons lacking Fyn. Increased protein tyrosine phosphor-
ylation following pervanadate treatment of wild-type
Fig. 4. Pro216 in tau is important for its interaction with Fyn-SH3.
CHO cells were transiently cotransfected with Fyn and V5-tagged
wild-type (WT) or mutant P216A or P233A tau. (A) CHO cell lysates
and proteins pulled down by GST–Fyn-SH3 beads were probed
with antibodies against tau (V5) and GST. Numbers on the left indi-
cate molecular masses (kDa). (B) Bar chart showing the proportion
of total tau pulled down by GST–Fyn-SH3 beads. Values shown are
mean fold change from control ± SEM. N =6.*P < 0.05.
A. Usardi et al. Tyrosine phosphorylation-dependent tau–Fyn binding
FEBS Journal 278 (2011) 2927–2937 ª 2011 The Authors Journal compilation ª 2011 FEBS 2931
and Fyn-deficient neuronal cultures was detected with
the phosphotyrosine antibody 4G10 (Fig. 5). There
was no apparent decrease in 4G10 immunoreactivity in
homogenates from Fyn-deficient neurons, and this
probably reflects compensation for the loss of Fyn by
other Src family kinases, as has been previously dem-
onstrated [29].
A tau species of  50–55 kDa, corresponding to
endogenous mouse tau, was detected in lysates and
DRMs isolated from vehicle-treated wild-type and
Fyn-deficient neurons, confirming our previous find-

ings [9]. In homogenate from wild-type, but not Fyn-
deficient, neurons, pervanadate treatment increased the
density of an  55-kDa tau band, suggesting increased
Fyn-dependent phosphorylation of tau in response to
pervanadate in wild-type neurons (Fig. 5). Interest-
ingly, there was also an increase in the amount of
DRM-associated tau isolated from wild-type neurons
(Fig. 5A). In contrast, pervanadate did not appear to
induce any change in the amount of tau in the DRM
fraction isolated from Fyn-deficient neurons (Fig. 5B).
Quantitation of the amount of DRM-associated tau as
a proportion of the total tau in the corresponding cell
lysates revealed a significant increase in the amount of
tau present in DRMs from wild-type mice
(P < 0.001), but not from Fyn-deficient mice, when
compared with control (catalase-treated) neurons. This
finding suggests that increased tyrosine phosphoryla-
tion of cellular proteins leads to enhanced trafficking
of tau to DRMs, and that this process is mediated by
Fyn. As we also found that the tyrosine phosphoryla-
tion status of tau regulates its interactions with
Fyn-SH2, it is possible that interactions between tau
and Fyn-SH2 play an important role in regulating
the intracellular trafficking of tau to membrane
compartments.
Discussion
Interactions between tau and Fyn play a critical role
in governing neuronal responses to elevated Ab levels
in models of AD [8], suggesting that disruption of the
association of these proteins could represent a poten-

tial therapeutic strategy for the treatment of AD. Here,
we used GST fusion proteins of Fyn-SH2 and Fyn-
SH3 to further investigate the mechanisms by which
tau and Fyn interact. We determined that tau binds to
both Fyn-SH2 and Fyn-SH3, and only the former of
these associations is mediated by tyrosine phosphoryla-
tion. With the methods employed here, it was not pos-
sible to accurately quantify differences in the relative
proportions of tau capable of binding to Fyn-SH2 and
Fyn-SH3, as there may be variations in the affinity of
Fyn-SH2 and Fyn-SH3 for GST beads. However, our
data suggest that several-fold more tau binds to Fyn-
SH3 than to Fyn-SH2.
Tau binds predominantly to Fyn-SH3, which has a
specific affinity for PXXP motifs in proteins [20,21].
Tau binding to SH3 domains is regulated by the phos-
phorylation of tau on specific serine⁄ threonine residues
[21], and we show here that Fyn-SH3–tau interactions
are not influenced by the tyrosine phosphorylation sta-
tus of tau. Using specific tau constructs in which either
Pro216 or Pro233 was mutated to disrupt critical
PXXP motifs implicated in Fyn binding, we have
shown that Pro216 is especially important for the
interaction of tau with Fyn-SH3, in line with our pre-
vious work [21]. These findings, however, contrast with
those of a previous study in which a deletion mutant
of tau lacking residues 169–179 displayed a 90%
reduction in its binding to Fyn-SH3 [20]. The reason
for this discrepancy is unclear; however, it is possible
that the deletion mutants of tau used previously may

have altered the tau folding⁄ conformation, thus mask-
ing the interaction between P216A of tau and Fyn.
Fig. 5. Tyrosine phosphorylation influences tau content in neuronal
DRMs. Western blots of homogenates and DRMs isolated from
neuronal cultures prepared from (A) wild-type (WT) and (B) Fyn-defi-
cient (Fyn
) ⁄ )
) mice. Neurons were treated with pervanadate (P) or
catalase (C). Western blots were probed with antibodies against
total (phosphorylated and nonphosphorylated) endogenous tau
(Dako), the DRM marker flotillin-1, Fyn, or phosphotyrosine (4G10).
The arrow indicates an  55-kDa tau species that increases in den-
sity in response to pervanadate treatment of WT neurons. Num-
bers on the left indicate molecular masses (kDa). The bar charts
below show the amount of tau in DRM fractions as a proportion of
total tau in cell lysates, expressed as the fold change from control.
Values shown are mean ± SEM. N = 8. ***P < 0.005.
Tyrosine phosphorylation-dependent tau–Fyn binding A. Usardi et al.
2932 FEBS Journal 278 (2011) 2927–2937 ª 2011 The Authors Journal compilation ª 2011 FEBS
In the work presented here, P216A and P233A
mutant tau proteins were correctly synthesized and
expressed in CHO cells, and both of these proteins
were able to interact, but to differing extents, with
Fyn-SH3. This suggests that the results shown here are
not artefactual, owing to incorrect folding of the tau
mutants, at least not in the regions responsible for
SH3 binding. Indeed, previous work by others showed
that the association between Fyn and tau was reduced,
but still maintained, when a tau mutant lacking an
entire PXXP motif was used in similar experiments

[20]. We therefore conclude that Pro216 in tau plays
an important role in its binding to Fyn-SH3. However,
tau possesses seven PXXP motifs, five of which have
not been investigated, and we therefore cannot exclude
the possibility that other prolines in tau may be impor-
tant for its binding to Fyn-SH3.
Here, we have demonstrated, for the first time, that
tau interacts with Fyn-SH2. Proteins harbouring SH2
domains bind to phosphorylated tyrosines on their tar-
get proteins, thereby coupling the activity of tyrosine
kinases with intracellular signalling pathways [28]. In
our model system, in which exogenous tau and Fyn
were expressed in non-neuronal cells, we found that
only a small proportion of tau is associated with Fyn-
SH2, both under control conditions and following per-
vanadate treatment to induce tyrosine phosphoryla-
tion. The magnitude of the increased tyrosine
phosphorylation that we observed following pervana-
date treatment is similar to that observed following
treatment of cells with physiological amounts of Ab
[30], and thus appears to have physiological relevance.
The influence of this relatively small pool of Fyn-SH2-
bound tau on potential neuronal responses to neuro-
toxic insults such as Ab remains to be determined. The
use of mutant tau constructs in which individual tyro-
sines were mutated to phenylalanine allowed us to
demonstrate that the tyrosine phosphorylation status
of tau significantly influences its ability to bind Fyn-
SH2. Indeed, we determined that Tyr18, the tau resi-
due apparently preferred by Fyn [14,17,18], plays a

dominant role in mediating the association of tau with
Fyn-SH2.
Tau phosphorylated at Tyr18 has been detected in a
proportion of tangles in early AD brain [31], and in
paired helical filament tau extracted from AD brain
[17,26]. Furthermore, Tyr18 phosphorylated tau has
been found in DRMs following treatment of neuronal
cells with Ab [14], and we have previously shown that
tau trafficking to DRMs in response to Ab treatment
of primary neurons is Fyn-dependent [9]. Here, using
wild-type and Fyn-deficient neurons, we show that
Fyn mediates the tyrosine phosphorylation-induced
recruitment of tau to neuronal DRMs. These mem-
brane microdomains are widely recognized as hubs for
intracellular signalling [32,33], and changes in the pro-
tein composition of DRMs are associated with the
induction of Ab-induced neurotoxicity [9]. Ittner et al.
[11] have suggested that interactions between tau and
Fyn in dendrites play a critical role in mediating Ab-
induced neurotoxicity by influencing the stability of
NMDA receptor–PSD-95 complexes. Interestingly,
both NMDA receptors and PSD-95 shuttle between
DRMs and postsynaptic densities under certain condi-
tions, including during learning [34], and PSD-95 plays
an important role in positioning NMDA receptors in
DRMs [34]. Thus, it is conceivable that tau and Fyn
might exist in a complex with NMDA receptors and
PSD-95 in neurons. Activation of signalling pathways
that lead to increased Fyn activity could therefore
affect the tyrosine phosphorylation of tau, which

could potentially modulate complex formation, and
result in altered trafficking into neuronal membrane
compartments.
In summary, the results presented here suggest that
tyrosine phosphorylation mediates the association of
tau with Fyn-SH2, but not with Fyn-SH3. This dem-
onstrates that different molecular mechanisms exist for
these two distinct interactions of Fyn and tau, with
probably disparate downstream consequences. Further-
more, these results support the view that non-micro-
tubule associations of tau are important for normal
physiological function in neurons, and reinforce the
suggestion that tau is itself involved in intracellular sig-
nalling pathways ⁄ mechanisms. As its interaction with
Fyn is important for tau localization in neurons, regu-
lation of the cellular signalling function of this micro-
tubule-associated protein could also have significant
implications during the progression of neurodegenera-
tive diseases, such as AD, in which both tau and Fyn
are implicated.
Experimental procedures
Plasmids and cell transfection
A plasmid expressing either the longest isoform of human
CNS tau, containing two N-terminal inserts (2N) and four
microtubule-binding repeats (4R), 2N4R tau, was a
generous gift from M. Goedert (Medical Research Council
Laboratory of Molecular Biology, Cambridge, UK) [35].
Generation of 2N4R tau constructs, each with a single tyro-
sine replaced by phenylalanine (Y18F, Y29F, Y197F,
Y310F and Y394F), or all with five tyrosines replaced by

phenylalanines (YallF), has been described previously [25].
To generate tau constructs with individual prolines replaced
A. Usardi et al. Tyrosine phosphorylation-dependent tau–Fyn binding
FEBS Journal 278 (2011) 2927–2937 ª 2011 The Authors Journal compilation ª 2011 FEBS 2933
by alanines, a QuikChange XL site-directed mutagenesis
kit (Stratagene, Amsterdam, the Netherlands) was used.
The primers used were as follows: P216A forward primer,
5¢-ACC C CG TCC CT T GCA ACC CCA CCC ACC-3 ¢;
P216A reverse primer, 5¢-GGT GGG TGG GGT TGC AA
G GGA CGG GGT-3¢; P233A forward primer, 5¢-GCA G
TG GTC CGG ACT CCA GCC AAG TCG CCG-3¢ (Primer
B); and P233A reverse primer, 5¢-CGG CGA CTT GGC
TGG AGT CCG GAC CAC TGC-3¢. P216A and P233A
were generated by use of appropriate primers with the plas-
mid expressing wild-type tau as template DNA. The cDNA
sequence of the full insert was determined for each tau con-
struct. For expression in mammalian cells, cDNA encoding
tau was subcloned into the pcDNA3.1 ⁄ V5-His-TOPOvector
(Invitrogen, Paisley, UK), yielding a construct with C-termi-
nal V5 and His tags, as described previously [25]. Human
Fyn cDNA in a pSG5 vector was a gift from D. Markby
(Sugen, San Francisco, CA, USA). Constructs for the bacte-
rial expression of GST have been described previously [21].
Plasmids expressing Fyn-SH3 and Fyn-SH2 were obtained
from S. Anderson (University of Colorado Health Sciences
Center, Denver, CO, USA). GST–Fyn-SH2 and GST–Fyn-
SH3 were generated by subcloning Fyn-SH2 and Fyn-SH3
into the plasmid pGEX-5X-2.
CHO cell culture and treatment
CHO cells were maintained in DMEM supplemented with

10% (v ⁄ v) fetal bovine serum, 2 m
ML-glutamine, and
100 UÆmL
)1
penicillin ⁄ 100 lgÆmL
)1
streptomycin, and incu-
bated at 37 °Cina5%CO
2
atmosphere. Cells were plated
into six-well dishes and transfected 24 h later by using
Lipofectamine Plus Reagent (Invitrogen), following the
manufacturer’s instructions. Pervandate and catalase were
prepared as described previously [25]. Briefly, vanadate
stock solution was prepared as a 200 m
M solution of
sodium orthovanadate (pH 10). Pervanadate was prepared
as a ·100 stock by adding 50 lL of 200 m
M sodium ortho-
vanadate and 1.6 lL of 30% (w ⁄ w) hydrogen peroxide to
948.4 lL of water for 5 min at room temperature, giving
10 m
M sodium orthovanadate and 16.3 mM hydrogen per-
oxide. After 5 min at room temperature, the excess hydro-
gen peroxide was removed by addition of 200 lgÆmL
)1
catalase (520 UÆmL
)1
) and incubation for an additional
5 min, as described by Huyer et al. [27]. Twenty-four hours

after transfection, cells were treated with either 100 l
M per-
vanadate or 2 lgÆmL
)1
catalase (control) for 20 min, prior
to harvesting for western blots, as described below.
Preparation of GST fusion proteins and GST
pulldown
Generation of GST, GST–Fyn-SH2 and GST–Fyn-SH3
and subsequent coupling to glutathione beads have been
described previously [21]. Following CHO cell transfection
and ⁄ or cell treatments, cells were washed once with
NaCl ⁄ P
i
and harvested into ice-cold lysis buffer (25 mM
Tris ⁄ HCl, pH 7.5, containing 10% (v ⁄ v) glycerol, 0.5% (w ⁄ v)
Triton X-100) 1 m
M EDTA, 1 mM EGTA, 1 mM sodium
orthovanadate, 150 m
M sodium chloride, 10 mM sodium
fluoride, and complete protease inhibitor cocktail (minus
EDTA; Roche, Burgess Hill, UK), and allowed to solubi-
lize for 15 min on ice prior to centrifugation at 15 000 g for
5 min at 4 °C to remove insoluble debris. Following deter-
mination of protein concentration by bicinchoninic acid
protein assay (Thermo Scientific, Rockford, IL, USA),
equal amounts of protein were incubated with GST, GST–
Fyn-SH2 or GST–Fyn-SH3 beads for 1 h at 4 °C with
rotation. The beads were pelleted by centrifugation at 500 g
for 1 min at 4 °C, the supernatant was discarded, and the

beads washed three times with lysis buffer. Laemmli sample
buffer was added, and the samples were heated to 100 °C
for 5 min to release bound proteins. Proteins in CHO cell
lysates were separated on 10–12% polyacrylamide gels,
transferred to nitrocellulose, and probed with an antibody
to tau. The amount of tau in each sample was quantified
by densitometry, and samples were adjusted by dilution in
lysis buffer to ensure equal tau protein concentrations for
subsequent determination of the relative amounts of GST-
bound proteins.
Animals and culture of primary cortical neurons
Wild-type and Fyn-deficient (Fyn
) ⁄ )
) mice were obtained
from The Jackson Laboratory (Bar Habor, Maine, USA).
Fyn
) ⁄ )
mice were maintained on a mixed B6129F2 ⁄ J back-
ground, and the same strain was used to provide wild-type
controls. All animal work was licensed under the Animals
(Scientific Procedures) Act 1986, reviewed by the ethical
review panel of King’s College London, Institute of Psychia-
try and the Home Office inspectorate, and performed in
accordance with the European Communities Council Direc-
tive of 24 November 1986 (86 ⁄ 609 ⁄ EEC). Primary cortical
neurons were prepared from embryonic day 16 wild-type and
Fyn-deficient mouse embryos, as described previously [9].
Cells were plated onto six-well dishes coated with poly(
D-
lysine) (5 lgÆmL

)1
) at a density of 1 · 10
6
cells per well, and
cultured in Neurobasal medium (Invitrogen) containing
2% (v ⁄ v) B-27 serum-free supplement, 0.5 m
ML-glutamine,
100 UÆmL
)1
penicillin, and 100 lgÆmL
)1
streptomycin. Cells
were incubated at 37 °C in a 5% CO
2
atmosphere for 7 days
prior to use. To obtain total cell lysates from CHO cells and
primary cortical cultures, cells were washed in NaCl ⁄ Tris
and harvested in lysis buffer, as described above.
Isolation of DRMs
DRMs were isolated as described previously [36]. Briefly,
primary cortical neurons were washed in NaCl ⁄ Tris and
lysed in isolation buffer containing 1% (w ⁄ v) Chapso in
Tyrosine phosphorylation-dependent tau–Fyn binding A. Usardi et al.
2934 FEBS Journal 278 (2011) 2927–2937 ª 2011 The Authors Journal compilation ª 2011 FEBS
MBS buffer (25 mM Mes, 150 mM sodium chloride, pH 6.5)
containing 10 m
M magnesium chloride, 10 mM sodium fluo-
ride, 2 m
M sodium orthovanadate, 1 mM EGTA, and 5 mM
dithiothreitol. Lysates were homogenized by 20 strokes in a

Dounce homogenizer, and incubated on ice for 30 min.
One millilitre of homogenate was mixed with 1 mL of
90% (w ⁄ v) sucrose in MBS buffer, and placed in a 12-mL
ultracentrifuge tube. A discontinuous 5%–35%–45%
sucrose gradient was formed by layering 6 mL of
35% (w ⁄ v) sucrose in MBS buffer on top of the 2-mL
homogenate, followed by 4 mL of 5% (w ⁄ v) sucrose in
MBS buffer. Samples were centrifuged at 180 000 g for
18 h at 4 °C in a Beckman SW41 rotor (Beckman Instru-
ments, Fullerton, CA, USA). Twelve 1-mL fractions were
collected from the top of each gradient. To concentrate
DRMs in fractions 4–5, 10 mL of MBS buffer was added
and the samples were centrifuged at 110 000 g for 1 h at
4 °C in a Beckman SW41 rotor. The resulting pellet was
solubilized in 100 lLof20m
M Tris ⁄ HCl (pH 7.4) contain-
ing 8
M urea, 10 mM NaF, 2 mM Na
3
VO
4
and 5 mM dith-
iothreitol. Samples were mixed with Laemlli buffer and
heated at 100 °C for 5 min prior to analysis by SDS ⁄
PAGE.
SDS

PAGE and western blotting
Denaturing PAGE and western blotting were performed as
described previously [37]. Briefly, separated proteins were

blotted onto nitrocellulose membranes (Whatman, Maid-
stone, UK) and blocked in 5% (w ⁄ v) nonfat
milk ⁄ 0.05% (v ⁄ v) Tween-20 in NaCl ⁄ P
i
for 1 h. After block-
ing, membranes were incubated overnight at 4 °C in blocking
solution containing primary antibody. The antibodies used
were directed against tau (total tau, rabbit polyclonal; Dako,
Glostrup, Denmark), V5 (mouse monoclonal; Invitrogen),
phosphotyrosine (4G10, mouse monoclonal; Millipore, Bill-
erica, MA, USA), Fyn (mouse monoclonal; Santa Cruz Bio-
technology, Santa Cruz, CA, USA) and GST (mouse
monoclonal; GE Healthcare, Little Chalfont, UK). After
three washes in NaCl ⁄ P
i
containing 0.05% (v ⁄ v) Tween-20,
blots were incubated with the appropriate fluorophore-conju-
gated goat secondary antibody (IRDye800 goat anti-rabbit;
Rockland, Gilbertsville, PA, USA or Alexa Fluor 680 goat
anti-mouse; Invitrogen) for 1 h at ambient temperature. Pro-
teins were visualized with the Odyssey imaging system
(Li-Cor Biosciences, Cambridge, UK). Scanned images were
analysed with the public domain
IMAGEJ program (http://
www.rsb.info.nih.gov/ij/).
Statistics
Statistical analyses were performed by t-test or ANOVA
with
GRAPHPAD PRISM 5.0 (GraphPad Software Inc., La
Jolla, CA, USA). Data are presented as mean ± standard

error of the mean (SEM).
Acknowledgements
We thank the following for gifts of materials: M.
Goedert (MRC Laboratory of Molecular Biology,
Cambridge, UK) for tau 2N4R cDNA; S. Anderson
(University of Colorado Health Sciences Center) for
pGEX constructs; and D. Markby (Sugen, San Fran-
cisco, CA, USA) for human Fyn cDNA. This work
was supported by Alzheimer’s Research UK, the Alz-
heimer’s Society, and the Medical Research Council.
References
1 Brady RM, Zinkowski RP & Binder LI (1995) Presence
of tau in isolated nuclei from human brain. Neurobiol
Aging 16, 479–486.
2 Sultan A, Nesslany F, Violet M, Begard S, Loyens A,
Talahari S, Mansuroglu Z, Marzin D, Sergeant N, Hu-
mez S et al. (2011) Nuclear tau, a key player in neuro-
nal DNA protection. J Biol Chem 286, 4566–4575.
3 Brandt R, Leger J & Lee G (1995) Interaction of tau
with the neural plasma membrane mediated by tau’s
amino-terminal projection domain. J Cell Biol 131,
1327–1340.
4 Maas T, Eidenmuller J & Brandt R (2000) Interaction
of tau with the neural membrane cortex is regulated by
phosphorylation at sites that are modified in paired heli-
cal filaments. J Biol Chem 275, 15733–15740.
5 Pooler AM, Usardi A, Evans CJ, Philpott KL, Noble
W & Hanger DP (2011) Dynamic association of tau
with neuronal membranes is regulated by phosphoryla-
tion. Neurobiol Aging, doi: 10.1016/j.neurobiolaging.

2011.01.005 [published online 7 March 2011].
6 Hanger DP, Seereeram A & Noble W (2009) Mediators
of tau phosphorylation in the pathogenesis of Alzhei-
mer’s disease. Expert Rev Neurother 9 , 1647–1666.
7 Hanger DP, Anderton BH & Noble W (2009) Tau
phosphorylation: the therapeutic challenge for neurode-
generative disease. Trends Mol Med 15, 112–119.
8 Ittner LM & Gotz J (2011) Amyloid-beta and tau –
a toxic pas de deux in Alzheimer’s disease. Nat Rev
Neurosci 12, 65–72.
9 Williamson R, Usardi A, Hanger DP & Anderton BH
(2008) Membrane-bound beta-amyloid oligomers are
recruited into lipid rafts by a fyn-dependent mechanism.
FASEB J 22, 1552–1559.
10 Williamson R, Scales T, Clark BR, Gibb G, Reynolds
CH, Kellie S, Bird IN, Varndell IM, Sheppard PW,
Everall I et al. (2002) Rapid tyrosine phosphorylation
of neuronal proteins including tau and focal adhesion
kinase in response to amyloid-beta peptide exposure:
involvement of Src family protein kinases. J Neurosci
22, 10–20.
11 Ittner LM, Ke YD, Delerue F, Bi M, Gladbach A, van
Eersel J, Wolfing H, Chieng BC, Christie MJ, Napier
A. Usardi et al. Tyrosine phosphorylation-dependent tau–Fyn binding
FEBS Journal 278 (2011) 2927–2937 ª 2011 The Authors Journal compilation ª 2011 FEBS 2935
IA et al. (2010) Dendritic function of tau mediates amy-
loid-beta toxicity in Alzheimer’s disease mouse models.
Cell 142, 387–397.
12 Roberson ED, Halabisky B, Yoo JW, Yao J, Chin J,
Yan F, Wu T, Hamto P, Devidze N, Yu GQ et al.

(2011) Amyloid-beta ⁄ Fyn-induced synaptic, network,
and cognitive impairments depend on tau levels in mul-
tiple mouse models of Alzheimer’s disease. J Neurosci
31, 700–711.
13 Rapoport M, Dawson HN, Binder LI, Vitek MP &
Ferreira A (2002) Tau is essential to beta-amyloid-
induced neurotoxicity. Proc Natl Acad Sci USA 99,
6364–6369.
14 Hernandez P, Lee G, Sjoberg M & Maccioni RB (2009)
Tau phosphorylation by cdk5 and Fyn in response to
amyloid peptide Abeta (25–35): involvement of lipid
rafts. J Alzheimers Dis 16, 149–156.
15 Chin J, Palop JJ, Yu GQ, Kojima N, Masliah E &
Mucke L (2004) Fyn kinase modulates synaptotoxicity,
but not aberrant sprouting, in human amyloid precur-
sor protein transgenic mice. J Neurosci 24, 4692–4697.
16 Chin J, Palop JJ, Puolivali J, Massaro C, Bien-Ly N,
Gerstein H, Scearce-Levie K, Masliah E & Mucke L
(2005) Fyn kinase induces synaptic and cognitive
impairments in a transgenic mouse model of Alzhei-
mer’s disease. J Neurosci 25, 9694–9703.
17 Lee G, Thangavel R, Sharma VM, Litersky JM, Bhas-
kar K, Fang SM, Do LH, Andreadis A, Van HG &
Ksiezak-Reding H (2004) Phosphorylation of tau by
fyn: implications for Alzheimer’s disease. J Neurosci 24,
2304–2312.
18 Lebouvier T, Scales TM, Williamson R, Noble W, Duy-
ckaerts C, Hanger DP, Reynolds CH, Anderton BH &
Derkinderen P (2009) The microtubule-associated pro-
tein tau is also phosphorylated on tyrosine. J Alzhei-

mers Dis 18, 1–9.
19 Scales TM, Derkinderen P, Leung KY, Byers HL,
Ward MA, Price C, Bird IN, Perera T, Kellie S,
Williamson R et al. (2011) Tyrosine phosphorylation of
tau by the SRC family kinases lck and fyn. Mol
Neurodegener 6, 12.
20 Lee G, Newman ST, Gard DL, Band H & Pancha-
moorthy G (1998) Tau interacts with src-family non-
receptor tyrosine kinases. J Cell Sci 111 (Pt 21),
3167–3177.
21 Reynolds CH, Garwood CJ, Wray S, Price C, Kellie S,
Perera T, Zvelebil M, Yang A, Sheppard PW, Varndell
IM et al. (2008) Phosphorylation regulates tau interac-
tions with Src homology 3 domains of phosphatidylino-
sitol 3-kinase, phospholipase Cgamma1, Grb2, and Src
family kinases. J Biol Chem 283, 18177–18186.
22 Anderson D, Koch CA, Grey L, Ellis C, Moran MF &
Pawson T (1990) Binding of SH2 domains of phospholi-
pase C gamma 1, GAP, and Src to activated growth
factor receptors. Science 250, 979–982.
23 Pleiman CM, Clark MR, Gauen LK, Winitz S, Cogge-
shall KM, Johnson GL, Shaw AS & Cambier JC (1993)
Mapping of sites on the Src family protein tyrosine kin-
ases p55blk, p59fyn, and p56lyn which interact with the
effector molecules phospholipase C-gamma 2, microtu-
bule-associated protein kinase, GTPase-activating pro-
tein, and phosphatidylinositol 3-kinase. Mol Cell Biol
13, 5877–5887.
24 Gotoh H, Okumura N, Yagi T, Okumura A, Shima T
& Nagai K (2006) Fyn-induced phosphorylation of

beta-adducin at tyrosine 489 and its role in their subcel-
lular localization. Biochem Biophys Res Commun 346,
600–605.
25 Derkinderen P, Scales TM, Hanger DP, Leung KY,
Byers HL, Ward MA, Lenz C, Price C, Bird IN, Perera
T et al. (2005) Tyrosine 394 is phosphorylated in Alz-
heimer’s paired helical filament tau and in fetal tau with
c-Abl as the candidate tyrosine kinase. J Neurosci
25,
6584–6593.
26 Bhaskar K, Hobbs GA, Yen SH & Lee G (2010) Tyro-
sine phosphorylation of tau accompanies disease pro-
gression in transgenic mouse models of tauopathy.
Neuropathol Appl Neurobiol 36, 462–477.
27 Huyer G, Liu S, Kelly J, Moffat J, Payette P, Kennedy
B, Tsaprailis G, Gresser MJ & Ramachandran C (1997)
Mechanism of inhibition of protein-tyrosine phosphata-
ses by vanadate and pervanadate. J Biol Chem 272,
843–851.
28 Pawson T (2004) Specificity in signal transduction: from
phosphotyrosine–SH2 domain interactions to complex
cellular systems. Cell 116, 191–203.
29 Smith CL, Mittaud P, Prescott ED, Fuhrer C & Burden
SJ (2001) Src, Fyn, and Yes are not required for neuro-
muscular synapse formation but are necessary for sta-
bilization of agrin-induced clusters of acetylcholine
receptors. J Neurosci 21, 3151–3160.
30 Luo YQ, Hirashima N, Li YH, Alkon DL, Sunderland
T, Etcheberrigaray R & Wolozin B (1995) Physiological
levels of beta-amyloid increase tyrosine phosphorylation

and cytosolic calcium. Brain Res 681 , 65–74.
31 Horowitz PM, Patterson KR, Guillozet-Bongaarts AL,
Reynolds MR, Carroll CA, Weintraub ST, Bennett
DA, Cryns VL, Berry RW & Binder LI (2004) Early
N-terminal changes and caspase-6 cleavage of tau in
Alzheimer’s disease. J Neurosci 24, 7895–7902.
32 Romanelli RJ & Wood TL (2008) Directing traffic in
neural cells: determinants of receptor tyrosine kinase
localization and cellular responses. J Neurochem 105,
2055–2068.
33 Lingwood D & Simons K (2010) Lipid rafts as a mem-
brane-organizing principle. Science 327, 46–50.
34 Delint-Ramirez I, Salcedo-Tello P & Bermudez-Rattoni
F (2008) Spatial memory formation induces recruitment
of NMDA receptor and PSD-95 to synaptic lipid rafts.
J Neurochem 106, 1658–1668.
Tyrosine phosphorylation-dependent tau–Fyn binding A. Usardi et al.
2936 FEBS Journal 278 (2011) 2927–2937 ª 2011 The Authors Journal compilation ª 2011 FEBS
35 Smith MJ, Crowther RA & Goedert M (2000) The nat-
ural osmolyte trimethylamine N-oxide (TMAO) restores
the ability of mutant tau to promote microtubule
assembly. FEBS Lett 484, 265–270.
36 Williamson R, Thompson AJ, Abu M, Hye A, Usardi
A, Lynham S, Anderton BH & Hanger DP (2010) Iso-
lation of detergent resistant microdomains from cul-
tured neurons: detergent dependent alterations in
protein composition. BMC Neurosci 11, 120.
37 Kelleher I, Garwood C, Hanger DP, Anderton BH &
Noble W (2007) Kinase activities increase during the
development of tauopathy in htau mice. J Neurochem

103, 2256–2267.
A. Usardi et al. Tyrosine phosphorylation-dependent tau–Fyn binding
FEBS Journal 278 (2011) 2927–2937 ª 2011 The Authors Journal compilation ª 2011 FEBS 2937

×