You are on page 1of 9

JOURNAL OF NEUROCHEMISTRY | 2012 | 120 (Suppl. 1) | 140–148 doi: 10.1111/j.1471-4159.2011.07506.

Center for Neural Science, New York University, New York, New York, USA

Abstract dysfunction and memory deficits, and how these mechanisms


Mounting evidence suggests that amyloid beta-induced could provide novel avenues for therapeutic intervention to
impairments in synaptic plasticity that is accompanied by treat this devastating neurodegenerative disease.
cognitive decline and dementia represent key pathogenic Keywords: amyloid b, glycogen synthase kinase-3, long-term
steps of Alzheimer’s disease. In this study, we review recent potentiation, mammalian target of rapamycin, NMDA recep-
advances in the study of the molecular and cellular mecha- tors, reactive oxygen species.
nisms underlying Alzheimer’s disease-associated synaptic J. Neurochem. (2012) 120 (Suppl. 1), 140–148.

In April 1906, Auguste Deter, the first described Alzheimer familial AD cases, of which a significant portion carry either
patient, died, five years after she first was examined by APP or presenilin (PS, part of the c-secretase complex)
Dr Alois Alzheimer. With state of the art staining techniques in mutations, resulting in unusually high Ab levels and early
use at that time, Dr Alzheimer and his colleagues were able onset of dementia, which in most regards is identical to
to examine the brain pathology of Mrs Deter and identified sporadic AD (Selkoe 1998; Chapman et al. 2001). Further-
the now well-known, characteristic ‘plaques’. However, what more, most, if not all, transgenic mice that have been utilized
these plaques were composed of remained a mystery until to model AD and to evaluate potential therapeutic interven-
eight decades later when a sticky peptide called amyloid beta tions have included human APP and/or PS mutations because
(Ab) was purified and sequenced with modern biochemical these mice replicate AD-like amyloid deposition and age-
methods (Glenner and Wong 1984; Masters et al. 1985). The related, progressive memory impairment (Ashe 2001; Janus
discovery of Ab launched a new era of research on and Westaway 2001; Zahs and Ashe 2010).
Alzheimer’s disease (AD) and a new framework to explain Another piece of compelling evidence of Ab being a
AD pathogenesis. AD is the most common form of dementia causative factor in AD comes from studies in which synthetic
and gradually gets worse over time, with aging being the Ab was injected directly into various brain areas of animals,
highest risk factor (Querfurth and LaFerla 2010). In this primarily the hippocampus. In these studies, microinjection
review, we focus our discussion on the role of Ab in of the Ab peptide impaired working memory in a manner
impairments in synaptic plasticity and memory function. Our consistent with this type of memory deficit in AD patients
objective is to discuss recent advances in the study of the (McDonald et al. 1994; Cleary et al. 1995; Stéphan et al.
molecular and cellular mechanisms underlying AD-associ- 2001). Given that Ab by nature is a ‘sticky’ peptide that
ated synaptic dysfunction and memory impairment, and how tends to aggregate, an interesting question has arisen as to
these mechanisms could provide novel avenues for thera-
peutic intervention in this devastating disease of memory.
Received July 1, 2011; revised manuscript received September 8, 2011;
accepted September 20, 2011.
Ab-induced memory impairment Address correspondence and reprint requests to Eric Klann, PhD,
Center for Neural Science, New York University, 4 Washington Place,
It has been revealed that Ab is derived from amyloid Room 809, New York, NY 10003, USA. E-mail: eklann@cns.nyu.edu
precursor protein (APP) via abnormal sequential cleavage by Abbreviations used: AD, Alzheimer’s disease; APP, amyloid precur-
sor protein; GSK3, glycogen synthase kinase-3; LFS, low-frequency
b- and c-secretases (Kang et al. 1987; Esch et al. 1990; stimulation; LTD, long-term depression; LTP, long-term potentiation;
Selkoe 1998). Strong support for the pathogenic role of Ab mTOR, mammalian target of rapamycin; NMDAR, NMDA receptor; PS,
in AD first arose with breakthrough genetic findings in presenilin; ROS, reactive oxygen species; SOD, superoxide dismutase.

Ó 2011 The Authors


140 Journal of Neurochemistry Ó 2011 International Society for Neurochemistry, J. Neurochem. (2012) 120 (Suppl. 1), 140–148
Ab-induced synaptic and memory deficits in AD | 141

what assembled forms of Ab are actually responsible for the


observed memory disruption in AD. A series of studies
utilizing synthetic, natural, and human AD-derived Ab have
indicated that soluble Ab oligomers, with dimers being the
smallest species, are both necessary and sufficient to disrupt
normal learning and memory function (Cleary et al. 2005;
Lesné et al. 2006; Shankar et al. 2008).

Ab abrogates synaptic plasticity


Persistent change in neuronal circuits, or synaptic plasticity,
is widely thought to be required for learning and memory.
One well-known and intensely studied form of synaptic
plasticity at hippocampal synapses is long-term potentiation
(LTP), broadly defined as an activity-dependent increase in
synaptic strength, which is measured by the slope (and/or the
amplitude) of the excitatory post-synaptic potential in a
neuron after either a brief burst of stimulation delivered to
pre-synaptic afferents or following various pharmacological
treatments. Ever since it was described more than 40 years
ago (Bliss and Lomo 1973), LTP has remained the leading
cellular and synaptic model for learning and memory (Bliss
and Collingridge 1993; Malenka and Nicoll 1999; Kandel
2001; Malenka 2003). A plethora of studies have been
conducted to examine the effects of Ab on LTP in vitro and
in vivo. In most cases, LTP can be blocked by either direct
exogenous Ab application (at a concentration of 100 nM or
higher for synthetic Ab) (Fig. 1b), or in AD transgenic
mouse models in which an abnormally high levels of Ab are
present (Rowan et al. 2005). It is interesting to note that
multiple lines of evidence suggest that there is an early, pre-
plaque phase when learning and memory deficits are not
detected in AD transgenic mice, but LTP is already impaired
(Oddo et al. 2003; Jacobsen et al. 2006; Ma et al. 2010)
(Fig. 1c). In addition to the possibility that the behavioral
paradigms being used currently are not sufficiently sensitive
to detect small, early changes in memory function, these Fig. 1 Ab impairs hippocampal long-term potentiation. (a) Schaffer
observations are in line with the hypothesis that soluble Ab collateral efferents are stimulated to elicit a field excitatory post-syn-
aptic potential (fEPSP) at synapses in the dendritic layer of area CA1
oligomers, but not plaque cores, in AD are synaptotoxic
in acute hipocampal slices. (b) Application of exogenous synthetic
(Haass and Selkoe 2007; Querfurth and LaFerla 2010).
Ab1-42 (500 nM) results in the blockade of long-term potentiation
Interestingly, in clinical studies a group of patients has been
(LTP). (c) Hippocampal LTP is impaired in slices from the Tg2576
described with a very subtle memory syndrome, but they do transgenic mouse model of Alzheimer’s disease at 3–4 months of age,
not display dementia. These patients, who otherwise have which is before the onset of memory impairments and the formation of
normal cognition, are diagnosed as having minimal (or mild) amyloid plaques. Adapted from Ma et al. (2010).
cognitive impairment, often considered the harbinger of AD
(Selkoe 2004; Querfurth and LaFerla 2010). Thus, these
clinical observations are consistent with the basic experi- Singer 1993; Stanton 1996; Malenka and Bear 2004). It was
mental findings that have demonstrated that hippocampal reported that LTD in vivo induced by low-frequency
LTP impairments appear earlier than behavioral deficits in stimulation (LFS) can be facilitated by injection of different
AD transgenic mice. forms of synthetic Ab (Kim et al. 2001; Cheng et al. 2009).
Compared with LTP, much fewer studies have been Additional studies on LTD in vitro have confirmed that LFS-
conducted to examine the effects of Ab on long-term induced LTD is enhanced by Ab from various sources,
depression (LTD), another form of synaptic plasticity, which including synthetic, cell culture, and human AD brains
is a persistent decrease in synaptic efficacy (Artola and extracts (Shankar et al. 2008; Li et al. 2009). It should be

Ó 2011 The Authors


Journal of Neurochemistry Ó 2011 International Society for Neurochemistry, J. Neurochem. (2012) 120 (Suppl. 1), 140–148
142 | T. Ma and E. Klann

noted that it also has been reported that LTD induced by


stronger LFS protocol is not altered by Ab (Wang et al.
2002; Raymond et al. 2003).
Although numerous reports indicate that Ab causes
impaired synaptic plasticity, there are paradoxical lines of
evidence from in vitro and in vivo studies that have shown
that increased synaptic activity per se induces Ab secretion
(Kamenetz et al. 2003; Cirrito et al. 2005, 2008). Based on
these reports it has been speculated that though excessive
production of Ab is synaptotoxic, at lower concentrations Ab
may actually serve as a physiological molecule that regulates
normal synaptic plasticity and memory. Indeed, recent
studies indicate not only that endogenous Ab is indispens-
able for normal learning and memory, but also that Ab at
very low concentrations (picomolar) enhances hippocampal
LTP and memory formation (Puzzo et al. 2008; Garcia-Osta
and Alberini 2009).
Another interesting idea has been proposed to address the
‘Ab/synaptic activity paradox’ based on the theory of Fig. 2 Signaling pathways that have been demonstrated to contribute
intraneuronal Ab. Classically viewed as only being located to Ab-induced impairments in hippocampal long-term potentiation.
extracellularly, mounting evidence from AD transgenic mice
and human patients has pointed to the presence of Ab and subunit composition of synaptic NMDARs constantly
intraneuronally that may be involved in disease progression change during development and in response to neuronal
(Gouras et al. 2005; LaFerla et al. 2007). Based on findings activity and sensory experience. Indeed, activity-dependent,
that intraneuronal Ab is reduced by synaptic activation, bidirectional regulation of delivery and targeting of
whereas it is increased by synaptic inhibition, it has been NMDARs to synapses is known to play a pivotal role in
proposed that the pool of intracellular and extracellular Ab is synaptic plasticity (LTP and LTD), and in learning and
linked dynamically, and that it is the accumulation of memory (Lau and Zukin 2007; Lau et al. 2009; Rebola et al.
intraneuronal Ab that initiates its synaptotoxic effects 2010; Gladding and Raymond 2011). It has been argued that
(Tampellini et al. 2009, 2010; Gouras et al. 2010). either too little or too much NMDAR activity is harmful to
neurons (Hardingham and Bading 2003). Ab has been shown
to interfere with normal NMDAR trafficking by triggering
Deciphering the cellular mechanisms of Ab-
receptor internalization, thus reducing the number of surface
induced synaptic dysfuction
NMDARs (Snyder et al. 2005). Later, it was demonstrated
Developing a mechanistic understanding of the ability of Ab that Ab oligomers reduce NMDAR-mediated Ca2+ influx
to interfere with synaptic plasticity and memory could yield into active dendritic spines (Shankar et al. 2007). Further-
important insights into the pathophysiology of AD. During the more, a recent study suggests that depletion of the receptor
past ten years, many groups have carried out elegant studies tyrosine kinase EphB2 mediates Ab-induced NMDAR
exploring the potential molecular and cellular signaling blockade (Cissé et al. 2011).
mechanisms underlying the synaptotoxic effects of Ab. In Though most studies on NMDARs have focused on
the following sections, we will mainly discuss advancements synaptic NMDARs that are largely located at the post-
in studying the role of NMDA receptors (NMDARs), synaptic density, NMDARs also exist outside synapses, and
mitochondrial reactive oxygen species (ROS), glycogen these extrasynaptic NMDARs likely have distinct roles in
synthase kinase-3 (GSK3), and the mammalian target of signaling transduction and gene regulation (Hardingham
rapamycin (mTOR) signaling pathways in mediating the toxic et al. 2002; Lau and Zukin 2007). Recently, it has been
effects of Ab on synaptic function. Remarkably, even though reported that over-activation of extrasynaptic NR2B-contain-
these mechanisms are often explored separately in AD ing NMDARs may be involved in Ab-induced LTD
research, it is possible that they are interconnected based on enhancement and LTP inhibition (Li et al. 2009, 2011).
current understanding of cell signaling pathways (Fig. 2). Interestingly, it also was shown that memantine, an NMDAR
antagonist and FDA-approved drug for AD treatment,
preferentially blocks extrasynaptic rather than synaptic
NMDA receptors
NMDAR-mediated currents (Xia et al. 2010), providing
NMDA receptors are glutamate-gated, heteromeric ion further evidence for a role of extrasynaptic NMDARs in the
channels that are highly permeable to Ca2+. The number pathophysiology of AD.

Ó 2011 The Authors


Journal of Neurochemistry Ó 2011 International Society for Neurochemistry, J. Neurochem. (2012) 120 (Suppl. 1), 140–148
Ab-induced synaptic and memory deficits in AD | 143

versely, it has been shown that over-expression of SOD-2 in


Mitochondrial ROS two different AD mouse models is capable of reducing brain
The role of ROS in synaptic plasticity and memory has been Ab deposition and preventing memory deficits (Dumont
described as a ‘double-edged sword’ (Massaad and Klann et al. 2009; Massaad et al. 2009). More recently, it was
2011). On one hand, ROS have a physiological role, in that demonstrated using both pharmacological and genetic
their production is necessary to maintain normal synaptic approaches that AD-associated synaptic plasticity impair-
plasticity, probably via activation of certain key signaling ments can be prevented and reversed by targeting ROS
molecules such as protein kinase C and mitogen-activated derived from mitochondria, but not NADPH oxidase (Ma
protein kinases (Bindokas et al. 1996; Klann 1998; Klann et al. 2011). An interesting implication that arises from these
et al. 1998; Knapp and Klann 2002; Kishida et al. 2005; and other studies is that ROS play different roles in synaptic
Huddleston et al. 2008). On the other hand, ROS have been plasticity, depending either on their subcellular localization
linked to synaptic pathology associated with aging and or on the age of the animals being studied (Kamsler and
neurodegenerative diseases, including AD (Balaban et al. Segal 2003; Hu et al. 2006, 2007). Such complexity of ROS
2005; Lin and Beal 2006). Indeed, recent investigations on function in the nervous system and their diverse effects on
brains of pre-clinical AD patients have provided evidence synaptic function may also help interpret the observation that
that oxidative stress is one of the earliest changes in AD current clinical trials with antioxidants such as vitamin E
pathogenesis (Zhu et al. 2007; Smith et al. 2010). Not resulted in either a marginally positive effect or no effect on
surprisingly, various types of ROS-mediated oxidative stress the cognitive function of AD patients (Praticò 2008).
are used consistently as biomarkers of AD brain pathology
(Praticò 2008). It is likely that in pathological situations, Glycogen synthase kinase-3
unusually high levels of ROS are produced constantly,
overwhelming the ability of the endogenous antioxidants, Two isoforms of GSK3 exist, GSK3a and GSK3b. The two
including superoxide dismutase (SOD) and vitamins E and C GSK isoforms are encoded by different genes, but are
to remove them, which then results in impairments in virtually identical in their catalytic regions (Woodgett 1990;
synaptic function (Fig. 3). For example, AD mutant mice Jope and Johnson 2004). For reasons that are not obvious,
with decreased mitochondrial SOD (SOD-2) expression the majority of studies have focused on GSK3b, and the
exhibit elevated levels of brain Ab and accelerated cognitive tissue-specific distribution, cellular localization, and func-
dysfunction (Li et al. 2004; Esposito et al. 2006). Con- tions of the two isoforms are controversial (Woodgett 1990;
Hoeflich et al. 2000; Asuni et al. 2006; Doble et al. 2007;
MacAulay et al. 2007; Force and Woodgett 2009). GSK3 is
unusual in that its constituitive activity is high and reduced
with phosphorylation (Ser 9 in GSK3b and Ser 21 in
GSK3a) by upstream kinases including Akt and Wnt (Hur
and Zhou 2010). Initially identified as a regulator of
glycogen metabolism (Embi et al. 1980), GSK3 is now
widely recognized as a critical component involved into
many cellular processes, and GSK3 dysfunction has been
implicated in major diseases (Frame and Cohen 2001; Jope
and Johnson 2004). Dysregulation of GSK3 activity has been
observed in both sporadic and familial AD cases. Many lines
of evidence point to a role of GSK3 in the development of
both classical neuropathological features of AD, including
amyloid plaques and neurofibrillary tangles that are mainly
composed of hyperphosphorylated tau (Jope and Johnson
2004; Hooper et al. 2008). Below, we will limit our
discussion on the interaction between GSK3 and Ab.
Fig. 3 Abnormal accumulation of Ab in Alzheimer’s disease induces
Increasing evidence suggests that the relationship between
overproduction of reactive oxygen species (ROS), such as superoxide
GSK3 and Ab is probably bidirectional. For example, high
anion, hydrogen peroxide, and hydroxyl radicals, which overwhelms
GSK3 activity may interfere with normal APP processing via
the ability of endogenous antioxidant systems (superoxide dismutate,
vitamins C and E, etc.) to effectively remove ROS. Consequently, the
modulation of the function of secretases, leading to Ab
role of ROS as physiological molecules in mediating normal synaptic accumulation (Sun et al. 2002; Phiel et al. 2003; Hooper
plasticity and memory becomes subordinate to their detrimental ef- et al. 2008). Suppression of GSK3 activity by either
fects as oxidative stressors, resulting in impairments of synaptic pharmacological inhibitors or genetic manipulations results
plasticity and memory. in abolishment of Ab production and reducing plaque

Ó 2011 The Authors


Journal of Neurochemistry Ó 2011 International Society for Neurochemistry, J. Neurochem. (2012) 120 (Suppl. 1), 140–148
144 | T. Ma and E. Klann

pathology in the brains of AD transgenic mice (Phiel et al. impairment of learning and memory function (Banko et al.
2003; Su et al. 2004). Interestingly, in one study the effects 2005, 2007; Parsons et al. 2006; Antion et al. 2008; Blundell
on APP cleavage and Ab production were specifically et al. 2008; Gafford et al. 2011; Hoeffer et al. 2011; Stoica
attributed to GSK3a and not GSK3b (Phiel et al. 2003). In et al. 2011).
contrast, Ab can activate GSK3 through dephosporylation, There is recent evidence suggesting a link between mTOR
which is linked to down-regulation of the PI3K/Akt signaling and AD, although the exact role that mTOR plays in AD is
pathway (Magrané et al. 2005; Peineau et al. 2008; Terwel controversial. The first controversy arises from a basic
et al. 2008; Lee et al. 2009; Jo et al. 2011). question: how is mTOR regulated (or dysregulated) in AD?
GSK3 also has been proposed to play an important role in Decreased mTOR signaling has been reported in the brains of
synaptic plasticity (Peineau et al. 2007, 2008) and evidence an APP-PS1 AD transgenic mouse model (Lafay-Chebassier
is accumulating to suggest the therapeutic potential of GSK3 et al. 2005), which is consistent with subsequent findings
inhibition for AD-related synaptic dysfunction. For example, that rapamycin, a specific inhibitor of mTORC1, exacerbates
it was reported that hippocampal LTP impairments in an AD neurotoxicity of Ab (Lafay-Chebassier et al. 2006). In
transgenic mouse could be rescued by two structurally contrast, up-regulation of mTOR signaling has been reported
distinct GSK3 antagonists (Ma et al. 2010). Moreover, it was in postmortem human AD brains, particularly in tangle-
reported recently that Ab-induced LTP failure can be bearing neurons (An et al. 2003). This finding is in
prevented by pre-treatment of brain slices with a specific agreement with a recent report showing that mTOR signaling
GSK3 inhibitor (Jo et al. 2011). Compared with the abun- is enhanced in the triple-transgenic mouse model of AD,
dance of evidence from in vitro studies linking GSK3 activity which in addition to APP and PS mutations, also contains a
to the pathogenesis of AD, much less is known about the role tau mutation (Caccamo et al. 2010). However, a more recent
of GSK3 in AD in vivo. A recent study examined the in vivo study with Tg2576 AD model mice revealed that mTOR
effects of lithium, an inhibitor of GSK3, on AD transgenic signaling is down-regulated at early ages, but not in the aged
mice and observed that although lithium was able to improve transgenic mice (Ma et al. 2010). Yet, another study of the
cognitive function at early stages, such ability was lost in PDAPP [also known as hAPP(J20)] AD transgenic mouse
aged AD transgenic mice (Fiorentini et al. 2010). Other new model reported no alteration in mTOR signaling (Spilman
and more specific small molecule inhibitors of GSK3 may et al. 2010). One possibility for the discrepancies among
have better potential than lithium as an AD therapy, though these studies is that each of them utilized a different AD
more investigations are needed to confirm this possibility mouse model. Therefore, it is possible that the regulation of
(Balaraman et al. 2006). mTOR by a mutation in PS and/or tau alters the response of
mTOR to excess Ab. This possibility seems likely because
mTOR is well known to integrate multiple signaling
Mammalian target of rapamycin
pathways in response to diverse stimuli, including stress
Mammalian target of rapamycin is an evolutionarily con- and inflammation (Reiling and Sabatini 2006; Hoeffer and
served serine/threonine protein kinase that plays an essential Klann 2010).
role in the control of mRNA translation and cell growth (Hay Given the diversity of findings on mTOR dysregulation in
and Sonenberg 2004; Averous and Proud 2006; Yang and AD, it should not be surprising that there is disagreement
Guan 2007). Interestingly, mTOR assembles into two distinct concerning whether mTOR should be targeted therapeuti-
complexes, mTORC1 and mTORC2, distinguished by dif- cally as a treatment for AD. This issue is complicated further
ferent binding proteins associated with mTOR and sensitivity because of critical studies on the association between the
to rapamycin (Bhaskar and Hay 2007; Huang and Manning mammalian aging process and the mTOR pathway that have
2009; Hoeffer and Klann 2010). To our knowledge, all shown that either feeding mice rapamycin or genetically
studies conducted thus far on AD have examined only deleting ribosomal S6 protein kinase 1 (S6K1), a down-
mTORC1 (referred as mTOR hereafter for the purpose of stream effector of mTOR, results in increased life span
simplicity). Numerous reports have firmly established a (Harrison et al. 2009; Selman et al. 2009). However,
pivotal role of mTOR in CNS function, including studies whether either hippocampal synaptic plasticity or hippocam-
that have shown that the activation of mTOR and the pus-dependent memory function was impacted in the animals
subsequent stimulation of translation initiation to boost was not addressed in these studies. Given the fact that aging
translational capacity is crucial for long-lasting synaptic is a well-established risk factor for AD, two groups recently
plasticity and the consolidation of long-term memory (Richter conducted studies where AD transgenic mice were fed
and Klann 2009; Hoeffer and Klann 2010). Moreover, rapamycin to inhibit mTOR signaling and found that
evidence linking mTOR to synaptic plasticity and long-term cognitive deficits could be rescued (Caccamo et al. 2010;
memory has been derived from studies using either pharma- Spilman et al. 2010). In contrast, around the same time it was
cological agents or genetically-modified mice, which have reported that Ab-induced impairments in synaptic plasticity
demonstrated that disruption of mTOR signaling results in could be reversed by up-regulating mTOR signaling via

Ó 2011 The Authors


Journal of Neurochemistry Ó 2011 International Society for Neurochemistry, J. Neurochem. (2012) 120 (Suppl. 1), 140–148
Ab-induced synaptic and memory deficits in AD | 145

either pharmacological methods or a genetic manipulation fibrillary pathology in Alzheimer’s disease. Am. J. Pathol. 163,
(Ma et al. 2010). 591–607.
Antion M. D., Merhav M., Hoeffer C. A., Reis G., Kozma S. C., Thomas
How does one reconcile these seemingly conflicting
G., Schuman E. M., Rosenblum K. and Klann E. (2008) Removal
findings with respect to mTOR, rapamycin, and AD? First, of S6K1 and S6K2 leads to divergent alterations in learning,
it should be noted that the mTOR pathway was manipulated memory, and synaptic plasticity. Learn. Mem. 15, 29–38.
in quite different ways (e.g. chronic vs. acute), and focused Artola A. and Singer W. (1993) Long-term depression of excitatory
on different aspects of mTOR function (e.g. autophagy- synaptic transmission and its relationship to long-term potentiation.
Trends Neurosci. 16, 480–487.
related Ab changes vs. as a signaling molecule in synaptic
Ashe K. H. (2001) Learning and memory in transgenic mice modeling
plasticity). Moreover, a similar paradox exists in AD research Alzheimer’s disease. Learn. Mem. 8, 301–308.
with respect to the insulin-PI3K-Akt pathway, which have Asuni A. A., Hooper C., Reynolds C. H., Lovestone S., Anderton B. H.
been well recognized as upstream regulators of mTOR. Ab and Killick R. (2006) GSK3alpha exhibits beta-catenin and tau
was shown to impair insulin-PI3K-Akt signaling (Magrané directed kinase activities that are modulated by Wnt. Eur. J.
Neurosci. 24, 3387–3392.
et al. 2005; Townsend et al. 2007; De Felice et al. 2009; Lee
Averous J. and Proud C. G. (2006) When translation meets transfor-
et al. 2009) and insulin treatment was reported to improve mation: the mTOR story. Oncogene 25, 6423–6435.
cognitive function in patients with early AD (Reger et al. Balaban R. S., Nemoto S. and Finkel T. (2005) Mitochondria, oxidants,
2008). In contrast, AD transgenic mice with reduced insulin and aging. Cell 120, 483–495.
signaling are protected against cognitive decline (Cohen Balaraman Y., Limaye A. R., Levey A. I. and Srinivasan S. (2006)
Glycogen synthase kinase 3beta and Alzheimer’s disease: patho-
et al. 2009). In summary, there is still significant disagree-
physiological and therapeutic significance. Cell. Mol. Life Sci. 63,
ment regarding the actual in vitro and in vivo role played by 1226–1235.
mTOR in AD, and for this reason further investigation is Banko J. L., Poulin F., Hou L., DeMaria C. T., Sonenberg N. and Klann
warranted before rapamycin should be considered as a E. (2005) The translation repressor 4E-BP2 is critical for eIF4F
treatment for individuals with AD. complex formation, synaptic plasticity, and memory in the hip-
pocampus. J. Neurosci. 25(42), 9581–9590.
Banko J. L., Merhav M., Stern E., Sonenberg N., Rosenblum K. and
Concluding remarks Klann E. (2007) Behavioral alterations in mice lacking the trans-
lation repressor 4E-BP2. Neurobiol. Learn. Mem. 87, 248–256.
With an aging population that continues to grow rapidly, the Bhaskar P. T. and Hay N. (2007) The two TORCs and Akt. Dev. Cell 12,
number of people afflicted with AD has been escalating 487–502.
Bindokas V. P., Jordan J., Lee C. C. and Miller R. J. (1996) Superoxide
considerably. Thus, AD has been proposed as the new
production in rat hippocampal neurons: selective imaging with
epidemic of the twenty-first century (Dyer et al. 2006). hydroethidine. J. Neurosci. 16, 1324–1336.
Given that there is still no available effective treatment for Bliss T. V. and Collingridge G. L. (1993) A synaptic model of memory:
AD, it is both necessary and urgent that neuroscientists long-term potentiation in the hippocampus. Nature 361, 31–39.
develop novel therapeutics. A multitude of studies in the past Bliss T. V. and Lomo T. (1973) Long-lasting potentiation of synaptic
transmission in the dentate area of the anaesthetized rabbit fol-
decade have illuminated AD as a disorder of synaptic
lowing stimulation of the perforant path. J. Physiol. 232, 331–356.
dysfunction due to the synaptotoxic effects of Ab. We look Blundell J., Kouser M. and Powell C. M. (2008) Systemic inhibition of
forward to the translation of the dysregulated moleclular mammalian target of rapamycin inhibits fear memory reconsoli-
signaling pathways we have discussed in this review and an dation. Neurobiol. Learn. Mem. 90, 28–35.
effective treatment for Alzheimer’s disease in the future. Caccamo A., Majumder S., Richardson A., Strong R. and Oddo S.
(2010) Molecular interplay between mammalian target of rapa-
mycin (mTOR), amyloid-beta, and Tau: effects on cognitive
impairments. J. Biol. Chem. 285, 13107–13120.
Acknowledgements
Chapman P. F., Falinska A. M., Knevett S. G. and Ramsay M. F. (2001)
This work was supported by National Institutes of Health grants Genes, models and Alzheimer’s disease. Trends Genet. 17, 254–
NS034007 and NS047834, and Alzheimer’s Association Investiga- 261.
tor. This review is dedicated to the memory of Dr. Mark A. Smith, a Cheng L., Yin W.-J., Zhang J.-F. and Qi J.-S. (2009) Amyloid beta-
protein fragments 25-35 and 31-35 potentiate long-term depression
great colleague and a better friend.
in hippocampal CA1 region of rats in vivo. Synapse 63, 206–214.
Cirrito J. R., Yamada K. A., Finn M. B., Sloviter R. S., Bales K. R., May
P. C., Schoepp D. D., Paul S. M., Mennerick S. and Holtzman D.
Conflict of interests M. (2005) Synaptic activity regulates interstitial fluid amyloid-beta
The authors declare no conflict of interests. levels in vivo. Neuron 48, 913–922.
Cirrito J. R., Kang J.-E., Lee J., Stewart F. R., Verges D. K., Silverio L.
M., Bu G., Mennerick S. and Holtzman D. M. (2008) Endocytosis
References is required for synaptic activity-dependent release of amyloid-beta
in vivo. Neuron 58, 42–51.
An W.-L., Cowburn R. F., Li L., Braak H., Alafuzoff I., Iqbal K., Iqbal Cissé M., Halabisky B., Harris J. et al. (2011) Reversing EphB2
I.-G., Winblad B. and Pei J.-J. (2003) Up-regulation of phos- depletion rescues cognitive functions in Alzheimer model. Nature
phorylated/activated p70 S6 kinase and its relationship to neuro- 469, 47–52.

Ó 2011 The Authors


Journal of Neurochemistry Ó 2011 International Society for Neurochemistry, J. Neurochem. (2012) 120 (Suppl. 1), 140–148
146 | T. Ma and E. Klann

Cleary J., Hittner J. M., Semotuk M., Mantyh P. and O’Hare E. (1995) Gouras G. K., Almeida C. G. and Takahashi R. H. (2005) Intraneuronal
Beta-amyloid(1-40) effects on behavior and memory. Brain Res. Ab accumulation and origin of plaques in Alzheimer’s disease.
682, 69–74. Neurobiol. Aging 26, 1235–1244.
Cleary J. P., Walsh D. M., Hofmeister J. J., Shankar G. M., Kuskowski Gouras G. K., Tampellini D., Takahashi R. H. and Capetillo-Zarate E.
M. A., Selkoe D. J. and Ashe K. H. (2005) Natural oligomers of (2010) Intraneuronal beta-amyloid accumulation and synapse
the amyloid-beta protein specifically disrupt cognitive function. pathology in Alzheimer’s disease. Acta Neuropathol. 119, 523–
Nat. Neurosci. 8, 79–84. 541.
Cohen E., Paulsson J. F., Blinder P. et al. (2009) Reduced IGF-1 sig- Haass C. and Selkoe D. J. (2007) Soluble protein oligomers in neu-
naling delays age-associated proteotoxicity in mice. Cell 139, rodegeneration: lessons from the Alzheimer’s amyloid beta-pep-
1157–1169. tide. Nat. Rev. Mol. Cell Biol. 8, 101–112.
De Felice F., Vieira M. N. N., Bomfim T. R., Decker H., Velasco P. Hardingham G. E. and Bading H. (2003) The Yin and Yang of NMDA
T., Lambert M. P., Viola K. L., Zhao W.-Q., Ferreira S. T. and receptor signalling. Trends Neurosci. 26, 81–89.
Klein W. L. (2009) Protection of synapses against Alzheimer’s- Hardingham G. E., Fukunaga Y. and Bading H. (2002) Extrasynaptic
linked toxins: insulin signaling prevents the pathogenic binding NMDARs oppose synaptic NMDARs by triggering CREB shut-off
of Abeta oligomers. Proc. Natl. Acad. Sci. USA 106, 1971– and cell death pathways. Nat. Neurosci. 5, 405–414.
1976. Harrison D. E., Strong R., Sharp Z. D. et al. (2009) Rapamycin fed late
Doble B. W., Patel S., Wood G. A., Kockeritz L. K. and Woodgett J. R. in life extends lifespan in genetically heterogeneous mice. Nature
(2007) Functional redundancy of GSK-3alpha and GSK-3beta in 460, 392–395.
Wnt/beta-catenin signaling shown by using an allelic series of Hay N. and Sonenberg N. (2004) Upstream and downstream of mTOR.
embryonic stem cell lines. Dev. Cell 12, 957–971. Genes Dev. 18, 1926–1945.
Dumont M., Wille E., Stack C., Calingasan N. Y., Beal M. F. and Lin M. Hoeffer C. A. and Klann E. (2010) mTOR signaling: at the crossroads of
T. (2009) Reduction of oxidative stress, amyloid deposition, and plasticity, memory and disease. Trends Neurosci. 33, 67–75.
memory deficit by manganese superoxide dismutase overexpres- Hoeffer C. A., Cowansage K. K., Arnold E. C. et al. (2011) Inhibition of
sion in a transgenic mouse model of Alzheimer’s disease. FASEB the interactions between eukaryotic initiation factors 4E and 4G
J. 23, 2459–2466. impairs long-term associative memory consolidation but not
Dyer M., Renner W. and Bachmann M. (2006) A second vaccine rev- reconsolidation. Proc. Natl. Acad. Sci. USA 108, 3383–3388.
olution for the new epidemics of the 21st century. Drug Discov. Hoeflich K. P., Luo J., Rubie E. A., Tsao M.-S., Jin O. and Woodgett J.
Today 11, 1028–1033. R. (2000) Requirement for glycogen synthase kinase-3beta in cell
Embi N., Rylatt D. and Cohen P. (1980) Glycogen synthase kinase-3 survival and NF-kappaB activation. Nature 406, 86–90.
from rabbit skeletal muscle. Separation from cyclic-AMP-depen- Hooper C., Killick R. and Lovestone S. (2008) The GSK3 hypothesis of
dent protein kinase and phosphorylase kinase. Eur. J. Biochem. Alzheimer’s disease. J. Neurochem. 104, 1433–1439.
107, 519–527. Hu D., Serrano F., Oury T. D. and Klann E. (2006) Aging-dependent
Esch F., Keim P., Beattie E., Blacher R., Culwell A., Oltersdorf T., alterations in synaptic plasticity and memory in mice that over-
McClure D. and Ward P. (1990) Cleavage of amyloid beta peptide express extracellular superoxide dismutase. J. Neurosci. 26, 3933–
during constitutive processing of its precursor. Science 248, 1122– 3941.
1124. Hu D., Cao P., Thiels E., Chu C. T., Wu G.-y., Oury T. D. and Klann E.
Esposito L., Raber J., Kekonius L., Yan F., Yu G.-Q., Bien-Ly N., (2007) Hippocampal long-term potentiation, memory, and lon-
Puoliväli J., Scearce-Levie K., Masliah E. and Mucke L. (2006) gevity in mice that overexpress mitochondrial superoxide dismu-
Reduction in mitochondrial superoxide dismutase modulates Alz- tase. Neurobiol. Learn. Mem. 87, 372–384.
heimer’s disease-like pathology and accelerates the onset of Huang J. and Manning B. (2009) A complex interplay between Akt,
behavioral changes in human amyloid precursor protein transgenic TSC2 and the two mTOR complexes. Biochem. Soc. Trans. 37,
mice. J. Neurosci. 26, 5167–5179. 217–222.
Fiorentini A., Rosi M. C., Grossi C., Luccarini I. and Casamenti F. Huddleston A. T., Tang W., Takeshima H., Hamilton S. L. and Klann E.
(2010) Lithium improves hippocampal neurogenesis, neuropa- (2008) Superoxide-induced potentiation in the hippocampus re-
thology and cognitive functions in APP mutant mice. PLoS ONE 5, quires activation of ryanodine receptor type 3 and ERK. J. Neu-
e14382. rophysiol. 99, 1565–1571.
Force T. and Woodgett J. R. (2009) Unique and overlapping functions of Hur E.-M. and Zhou F.-Q. (2010) GSK3 signalling in neural develop-
GSK-3 isoforms in cell differentiation and proliferation and car- ment. Nat. Rev. Neurosci. 11, 539–551.
diovascular development. J. Biol. Chem. 284, 9643–9647. Jacobsen J. S., Wu C.-C., Redwine J. M. et al. (2006) Early-onset
Frame S. and Cohen P. (2001) GSK3 takes centre stage more than behavioral and synaptic deficits in a mouse model of Alzheimer’s
20 years after its discovery. Biochem. J. 359, 1–16. disease. Proc. Natl. Acad. Sci. USA 103, 5161–5166.
Gafford G. M., Parsons R. G. and Helmstetter F. J. (2011) Consolidation Janus C. and Westaway D. (2001) Transgenic mouse models of Alz-
and reconsolidation of contextual fear memory requires mamma- heimer’s disease. Physiol. Behav. 73, 873–886.
lian target of rapamycin-dependent translation in the dorsal hip- Jo J., Whitcomb D. J., Olsen K. M. et al. (2011) Ab(1-42) inhibition of
pocampus. Neuroscience 182, 98–104. LTP is mediated by a signaling pathway involving caspase-3, Akt1
Garcia-Osta A. and Alberini C. M. (2009) Amyloid beta mediates and GSK-3b. Nat. Neurosci. 14, 545–547.
memory formation. Learn. Mem. 16, 267–272. Jope R. S. and Johnson G. V. W. (2004) The glamour and gloom of
Gladding C. M. and Raymond L. A. (2011) Mechanisms underlying glycogen synthase kinase-3. Trends Biochem. Sci. 29, 95–102.
NMDA receptor synaptic/extrasynaptic distribution and function. Kamenetz F., Tomita T., Hsieh H., Seabrook G., Borchelt D., Iwatsubo
Mol. Cell. Neurosci. [Epub ahead of print]. T., Sisodia S. and Malinow R. (2003) APP processing and synaptic
Glenner M. D. G. G. and Wong C. W. (1984) Alzheimer’s disease: initial function. Neuron 37, 925–937.
report of the purification and characterization of a novel cerebro- Kamsler A. and Segal M. (2003) Paradoxical actions of hydrogen per-
vascular amyloid protein. Biochem. Biophys. Res. Commun. oxide on long-term potentiation in transgenic superoxide dismu-
120(3), 885–890. tase-1 mice. J. Neurosci. 23, 10359–10367.

Ó 2011 The Authors


Journal of Neurochemistry Ó 2011 International Society for Neurochemistry, J. Neurochem. (2012) 120 (Suppl. 1), 140–148
Ab-induced synaptic and memory deficits in AD | 147

Kandel E. R. (2001) The molecular biology of memory storage: a dia- synaptic plasticity in a mouse model of Alzheimer’s disease. PLoS
logue between genes and synapses. Science 294, 1030–1038. ONE 5, e12845.
Kang J., Lemaire H.-G., Unterbeck A., Salbaum J. M., Masters C. L., Ma T., Hoeffer C. A., Wong H., Massaad C. A., Zhou P., Iadecola C.,
Grzeschik K.-H., Multhaup G., Beyreuther K. and Muller-Hill B. Murphy M. P., Pautler R. G. and Klann E. (2011) Amyloid
(1987) The precursor of Alzheimer’s disease amyloid A4 protein b-induced impairments in hippocampal synaptic plasticity are
resembles a cell-surface receptor. Nature 325, 733–736. rescued by decreasing mitochondrial superoxide. J. Neurosci. 31,
Kim J.-H., Anwyl R., Suh Y.-H., Djamgoz M. B. A. and Rowan M. J. 5589–5595.
(2001) Use-dependent effects of amyloidogenic fragments of MacAulay K., Doble B. W., Patel S., Hansotia T., Sinclair E. M.,
(beta)-amyloid precursor protein on synaptic plasticity in rat hip- Drucker D. J., Nagy A. and Woodget J. R. (2007) Glycogen
pocampus in vivo. J. Neurosci. 21, 1327–1333. synthase kinase 3alpha-specific regulation of murine hepatic gly-
Kishida K. T., Pao M., Holland S. M. and Klann E. (2005) NADPH cogen metabolism. Cell Metab. 6, 329–337.
oxidase is required for NMDA receptor-dependent activation of Magrané J., Rosen K. M., Smith R. C., Walsh K., Gouras G. K. and
ERK in hippocampal area CA1. J. Neurochem. 94, 299–306. Querfurth H. W. (2005) Intraneuronal beta-amyloid expression
Klann E. (1998) Cell-permeable scavengers of superoxide prevent long- downregulates the Akt survival pathway and blunts the stress re-
term potentiation in hippocampal area CA1. J. Neurophysiol. 80, sponse. J. Neurosci. 25(47), 10960–10969.
452–457. Malenka R. C. (2003) The long-term potential of LTP. Nat. Rev. Neu-
Klann E., Roberson E. D., Knapp L. T. and Sweatt J. D. (1998) A role rosci. 4, 923–926.
for superoxide in protein kinase C activation and induction of long- Malenka R. C. and Bear M. F. (2004) LTP and LTD: an embarrassment
term potentiation. J. Biol. Chem. 273, 4516–4522. of riches. Neuron 44, 5–21.
Knapp L. T. and Klann E. (2002) Potentiation of hippocampal synaptic Malenka R. C. and Nicoll R. A. (1999) Long-term potentiation – a
transmission by superoxide requires the oxidative activation of decade of progress? Science 285, 1870–1874.
protein kinase C. J. Neurosci. 22, 674–683. Massaad C. A. and Klann E. (2011) Reactive oxygen species in the
Lafay-Chebassier C., Paccalin M., Page G., Barc-Pain S., Perault-Pochat regulation of synaptic plasticity and memory. Antioxid. Redox
M. C., Gil R., Pradier L. and Hugon J. (2005) mTOR/p70S6k Signal. 14, 2013–2054.
signalling alteration by Abeta exposure as well as in APP-PS1 Massaad C. A., Washington T. M., Pautler R. G. and Klann E. (2009)
transgenic models and in patients with Alzheimer’s disease. Overexpression of SOD-2 reduces hippocampal superoxide and
J. Neurochem. 94, 215–225. prevents memory deficits in a mouse model of Alzheimer’s disease.
Lafay-Chebassier C., Pérault-Pochat M. C., Page G., Bilan A. R., Proc. Natl. Acad. Sci. USA 106, 13576–13581.
Damjanac M., Pain S., Houeto J.-L., Gil R. and Hugon J. (2006) Masters C. L., Simms G., Weinman N. A., Multhaup G., Mcdonald B. L.
The immunosuppressant rapamycin exacerbates neurotoxicity of and Beyreuther K. (1985) Amyloid plaque core protein in Alz-
Abeta peptide. J. Neurosci. Res. 84, 1323–1334. heimer disease and Down syndrome. Proc. Natl. Acad. Sci. USA
LaFerla F. M., Green K. N. and Oddo S. (2007) Intracellular amyloid- 82, 4245–4249.
beta in Alzheimer’s disease. Nat. Rev. Neurosci. 8, 499–509. McDonald M., Dahl E., Overmier J., Mantyh P. and Cleary J. (1994)
Lau C. G. and Zukin R. S. (2007) NMDA receptor trafficking in synaptic Effects of an exogenous beta-amyloid peptide on retention for
plasticity and neuropsychiatric disorders. Nat. Rev. Neurosci. 8, spatial learning. Behav. Neural Biol. 62, 60–67.
413–426. Oddo S., Caccamo A., Shepherd J. D., Murphy M. P., Golde T. E.,
Lau C. G., Takeuchi K., Rodenas-Ruano A., Takayasu Y., Murphy J., Kayed R., Metherate R., Mattson M. P., Akbari Y. and LaFerla F.
Bennett M. V. L. and Zukin R. S. (2009) Regulation of NMDA M. (2003) Triple-transgenic model of Alzheimer’s disease with
receptor Ca2 + signalling and synaptic plasticity. Biochem. Soc. plaques and tangles: intracellular Abeta and synaptic dysfunction.
Trans. 37, 1369–1374. Neuron 39, 409–421.
Lee H.-K., Kumar P., Fu Q., Rosen K. M. and Querfurth H. W. (2009) Parsons R. G., Gafford G. M. and Helmstetter F. J. (2006) Translational
The insulin/Akt signaling pathway is targeted by intracellular beta- control via the mammalian target of rapamycin pathway is critical
amyloid. Mol. Biol. Cell 20, 1533–1544. for the formation and stability of long-term fear memory in
Lesné S., Koh M. T., Kotilinek L., Kayed R., Glabe C. G., Yang A., amygdala neurons. J. Neurosci. 26, 12977–12983.
Gallagher M. and Ashe K. H. (2006) A specific amyloid-beta Peineau S., Taghibiglou C., Bradley C. et al. (2007) LTP inhibits LTD
protein assembly in the brain impairs memory. Nature 440, 352– in the hippocampus via regulation of GSK3beta. Neuron 53, 703–
357. 717.
Li F., Calingasan N. Y., Yu F. et al. (2004) Increased plaque burden in Peineau S., Bradley C., Taghibiglou C., Doherty A., Bortolotto Z. A.,
brains of APP mutant MnSOD heterozygous knockout mice. Wang Y. T. and Collingridge G. L. (2008) The role of GSK-3 in
J. Neurochem. 89, 1308–1312. synaptic plasticity. Br. J. Pharmacol. 153, S428–S437.
Li S., Hong S., Shepardson N. E., Walsh D. M., Shankar G. M. and Phiel C. J., Wilson C. A., Lee V. M.-Y. and Klein P. S. (2003) GSK-
Selkoe D. (2009) Soluble oligomers of amyloid Beta protein 3alpha regulates production of Alzheimer’s disease amyloid-beta
facilitate hippocampal long-term depression by disrupting neuronal peptides. Nature 423, 435–439.
glutamate uptake. Neuron 62, 788–801. Praticò D. (2008) Oxidative stress hypothesis in Alzheimer’s disease: a
Li S., Jin M., Koeglsperger T., Shepardson N. E., Shankar G. M. and reappraisal. Trends Pharmacol. Sci. 29, 609–615.
Selkoe D. J. (2011) Soluble A{beta} Oligomers Inhibit Long-Term Puzzo D., Privitera L., Leznik E., Fà M., Staniszewski A., Palmeri A.
Potentiation through a Mechanism Involving Excessive Activation and Arancio O. (2008) Picomolar amyloid-beta positively modu-
of Extrasynaptic NR2B-Containing NMDA Receptors. J. Neuro- lates synaptic plasticity and memory in hippocampus. J. Neurosci.
sci. 31, 6627–6638. 28, 14537–14545.
Lin M. T. and Beal M. F. (2006) Mitochondrial dysfunction and oxi- Querfurth H. W. and LaFerla F. M. (2010) Alzheimer’s disease. N. Engl.
dative stress in neurodegenerative diseases. Nature 443, 787–795. J. Med. 362, 329–344.
Ma T., Hoeffer C. A., Capetillo-Zarate E., Yu F., Wong H., Lin M. T., Raymond C. R., Ireland D. R. and Abraham W. C. (2003) NMDA
Tampellini D., Klann E., Blitzer R. D. and Gouras G. K. (2010) receptor regulation by amyloid-beta does not account for its inhi-
Dysregulation of the mTOR pathway mediates impairment of bition of LTP in rat hippocampus. Brain Res. 968, 263–272.

Ó 2011 The Authors


Journal of Neurochemistry Ó 2011 International Society for Neurochemistry, J. Neurochem. (2012) 120 (Suppl. 1), 140–148
148 | T. Ma and E. Klann

Rebola N., Srikumar B. N. and Mulle C. (2010) Activity-dependent Stoica L., Zhu P. J., Huang W., Zhou H., Kozma S. C. and Costa-
synaptic plasticity of NMDA receptors. J. Physiol. 588, 93–99. Mattioli M. (2011) Selective pharmacogenetic inhibition of mam-
Reger M., Watson G., Green P. et al. (2008) Intranasal insulin improves malian target of Rapamycin complex I (mTORC1) blocks long-
cognition and modulates beta-amyloid in early AD. Neurology 70, term synaptic plasticity and memory storage. Proc. Natl. Acad. Sci.
440–448. USA 108, 3791–3796.
Reiling J. H. and Sabatini D. M. (2006) Stress and mTORture signaling. Su Y., Ryder J., Li B. et al. (2004) Lithium, a common drug for bipolar
Oncogene 25, 6373–6383. disorder treatment, regulates amyloid-beta precursor protein pro-
Richter J. D. and Klann E. (2009) Making synaptic plasticity and cessing. Biochemistry 43, 6899–6908.
memory last: mechanisms of translational regulation. Genes Dev. Sun X., Sato S., Murayama O., Murayama M., Park J., Yamaguchi H.
23, 1–11. and Takashima A. (2002) Lithium inhibits amyloid secretion in
Rowan M. J., Klyubin I., Wang Q. and Anwyl R. (2005) Synaptic COS7 cells transfected with amyloid precursor protein C100.
plasticity disruption by amyloid beta protein: modulation by po- Neurosci. Lett. 321, 61–64.
tential Alzheimer’s disease modifying therapies. Biochem. Soc. Tampellini D., Rahman N., Gallo E. F. et al. (2009) Synaptic activity
Trans. 33, 563–567. reduces intraneuronal Abeta, promotes APP transport to synapses,
Selkoe D. J. (1998) The cell biology of beta-amyloid precursor protein and protects against Abeta-related synaptic alterations. J. Neurosci.
and presenilin in Alzheimer’s disease. Trends Cell Biol. 8, 447–453. 29, 9704–9713.
Selkoe D. J. (2004) Alzheimer disease: mechanistic understanding pre- Tampellini D., Capetillo-Zarate E., Dumont M., Huang Z., Yu F., Lin
dicts novel therapies. Ann. Intern. Med. 140, 627–638. M. T. and Gouras G. K. (2010) Effects of synaptic modulation
Selman C., Tullet J. M. A., Wieser D. et al. (2009) Ribosomal protein S6 on beta-amyloid, synaptophysin, and memory performance in
kinase 1 signaling regulates mammalian life span. Science 326, Alzheimer’s disease transgenic mice. J. Neurosci. 30, 14299–
140–144. 14304.
Shankar G. M., Bloodgood B. L., Townsend M., Walsh D. M., Selkoe D. Terwel D., Muyllaert D., Dewachter I., Borghgraef P., Croes S., Devijver
J. and Sabatini B. L. (2007) Natural oligomers of the Alzheimer H. and Leuven F. V. (2008) Amyloid activates GSK-3beta to
amyloid-beta protein induce reversible synapse loss by modulating aggravate neuronal tauopathy in bigenic mice. Am. J. Pathol. 172,
an NMDA-type glutamate receptor-dependent signaling pathway. 786–798.
J. Neurosci. 27, 2866–2875. Townsend M., Mehta T. and Selkoe D. J. (2007) Soluble Abeta inhibits
Shankar G. M., Li S., Mehta T. H. et al. (2008) Amyloid-beta protein specific signal transduction cascades common to the insulin
dimers isolated directly from Alzheimer’s brains impair synaptic receptor pathway. J. Biol. Chem. 282, 33305–33312.
plasticity and memory. Nat. Med. 14, 837–842. Wang H.-W., Pasternak J. F., Kuo H. et al. (2002) Soluble oligomers of
Smith M. A., Zhu X., Tabaton M. et al. (2010) Increased iron and free beta amyloid (1-42) inhibit long-term potentiation but not long-
radical generation in preclinical Alzheimer disease and mild cog- term depression in rat dentate gyrus. Brain Res. 924, 133–140.
nitive impairment. J. Alzheimers Dis. 19, 363–372. Woodgett J. R. (1990) Molecular cloning and expression of glycogen
Snyder E. M., Nong Y., Almeida C. G. et al. (2005) Regulation of NMDA synthase kinase-3/factor A. EMBO J. 9, 2431–2438.
receptor trafficking by amyloid-beta. Nat. Neurosci. 8, 1051–1058. Xia P., Huei-sheng Chen V., Zhang D. and Lipton S. A. (2010) Me-
Spilman P., Podlutskaya N., Hart M. J., Debnath J., Gorostiza O., Bre- mantine preferentially blocks extrasynaptic over synaptic NMDA
desen D., Richardson A., Strong R. and Galvan V. (2010) Inhi- receptor currents in hippocampal autapses. J. Neurosci. 30, 11246–
bition of mTOR by rapamycin abolishes cognitive deficits and 11250.
reduces amyloid-beta levels in a mouse model of Alzheimer’s Yang Q. and Guan K.-L. (2007) Expanding mTOR signaling. Cell Res.
disease. PLoS ONE 5, e9979. 17, 666–681.
Stanton P. K. (1996) LTD, LTP, and the sliding threshold for long-term Zahs K. R. and Ashe K. H. (2010) ‘Too much good news’ – are Alz-
synaptic plasticity. Hippocampus 6, 35–42. heimer mouse models trying to tell us how to prevent, not cure,
Stéphan A., Laroche S. and Davis S. (2001) Generation of aggregated Alzheimer’s disease? Trends Neurosci. 33, 381–389.
beta-amyloid in the rat hippocampus impairs synaptic transmission Zhu X., Su B., Wang X., Smith M. A. and Perry G. (2007) Causes of
and plasticity and causes memory deficits. J. Neurosci. 21, 5703– oxidative stress in Alzheimer disease. Cell. Mol. Life Sci. 64,
5714. 2202–2210.

Ó 2011 The Authors


Journal of Neurochemistry Ó 2011 International Society for Neurochemistry, J. Neurochem. (2012) 120 (Suppl. 1), 140–148

You might also like