You are on page 1of 12

Neuromol Med (2010) 12:1–12

DOI 10.1007/s12017-009-8104-z

REVIEW PAPER

An Overview of APP Processing Enzymes and Products


Vivian W. Chow • Mark P. Mattson •

Philip C. Wong • Marc Gleichmann

Received: 4 September 2009 / Accepted: 6 November 2009 / Published online: 24 November 2009
Ó US Government 2009

Abstract The generation of amyloid b-peptide (Ab) by Introduction


enzymatic cleavages of the b-amyloid precursor protein
(APP) has been at the center of Alzheimer’s disease (AD) Excess amyloid b-peptide (Ab) is believed to be a main
research. While the basic process of b- and c-secretase- contributor to the dysfunction and degeneration of neurons
mediated generation of Ab is text book knowledge, new that occurs in Alzheimer’s disease (AD) (see Thinakaran
aspects of Ab and other cleavage products have emerged in and Koo, 2008 for review). Ab is a 38–43 amino acid
recent years. Also our understanding of the enzymes peptide that is derived from the b-amyloid precursor pro-
involved in APP proteolysis has increased dramatically. All tein (APP) through sequential cleavages by b- and
of these discoveries contribute to a more complete under- c-secretase enzyme activities. The cleavage site for another
standing of APP processing and the physiologic and APP processing enzyme, a-secretase, lies within the Ab
pathologic roles of its secreted and intracellular protein sequence and, thus, precludes Ab formation. The amino-
products. Understanding APP processing is important for terminal fragment generated through a- or b-secretase is
any therapeutic strategy aimed at reducing Ab levels in called secreted APP (sAPP) a or b, respectively. The car-
AD. In this review, we provide a concise description of the boxy-terminal fragments (CTF) generated by a- and b-
current state of understanding the enzymes involved in secretase are called CTF83 and CTF99, respectively.
APP processing, the cleavage products generated by dif- c-Secretase cleavage of CTF83 and CTF99 will result in
ferent processing patterns, and the potential functions of the generation of p3 and Ab, respectively, as well as the
those cleavage products. amino-terminal APP intracellular domain (AICD) (Fig. 1).
a-Secretase activity is mediated by one or more enzymes
Keywords Amyloid beta  a-secretase  b-secretase  from the family of disintegrin and metalloproteinase
c-secretase  APP  AICD domain proteins (ADAM), with ADAM 9, 10, 17, and 19
being the most likely candidates (Fahrenholz et al. 2000;
Asai et al. 2003; Tanabe et al. 2006). Beta-site APP
cleaving enzyme 1 (BACE1) is the major b-secretase in the
brain (Vassar et al. 1999). A multiprotein complex con-
stitutes c–secretase activity. Four proteins are minimally
required for this complex: presenilin (PS) 1 or 2, nicastrin
V. W. Chow  P. C. Wong (Nct), presenilin enhancer 2 (Pen2), and anterior pharynx
Department of Pathology, Division of Neuropathology,
defective 1 (Aph-1) (Wolfe 2008).
The Johns Hopkins University School of Medicine,
Baltimore, USA Ab is at the center of AD drug development research, and
from this short introduction, it is clear that at least three paths
M. P. Mattson  M. Gleichmann (&) can be followed to reduce Ab production, namely inhibit b-
Laboratory of Neurosciences, National Institute on Aging,
or c-secretase, or enhance a-secretase activity. The picture
5th Floor Suite 100 BRC, 251 Bayview Blvd,
Baltimore, MD 21224, USA becomes more complex when subcellular localization
e-mail: gleichmannma@grc.nia.nih.gov of enzyme activities and amino acid modifications are
2 Neuromol Med (2010) 12:1–12

generally considered beneficial to neurons. Early in vitro


AD pathology
β Amyloid plaque Inflammation studies have demonstrated that sAPPa protects cultured
Oxidative stress
neurons against oxygen-glucose deprivation and excito-
fibrillization Neuron death toxicity by inhibiting calcium currents and increasing
potassium currents and, thus, stabilizing the resting mem-
LTP impairment brane potential (Mattson et al. 1993; Furukawa et al. 1996).
oligomerization

promotes neuronal survival,


binds to DR6/ neurite outgrowth, neural stem
sAPPa also promotes neurite outgrowth, synaptogenesis,
synaptic pruning
APP
cell proliferation, enhances LTP and cell adhesion (Mattson 1997; Gakhar-Koppole et al.
2008). The biologically relevant site for these actions was
sAPP β

located to the carboxy-terminus of sAPPa, spanning the

sAPPα
region from just amino-terminal to the b-secretase cleavage

site to the carboxy-terminal end with a heparin binding

P3
motif at the carboxy-terminus being of critical importance
β (Furukawa et al. 1996). The amino-terminal end of sAPPa
CTF99/CTF89

ex
α
CTF 83

γ γ was not required for these effects. In vivo studies reported


CTF83

PM
cyt that sAPPa, when administered intracerebroventricularly,
AICD50
AICD50

enhances learning and memory in mice and rats (Meziane


Amyloidogenic pathway Non-amyloidgenic pathway
et al. 1998; Taylor et al. 2008). Interestingly, the latter
study reported that this effect was accompanied by
? ? increased long-term potentiation (LTP) and enhanced
N-methyl D-aspartate (NMDA) receptor-mediated currents,
which is difficult to reconcile with the in vitro observations
AICD50

Tip60
nucleus Transcriptional activation of p53,
Fe65 GSK3β, Neprilysin, EGFR, (in vitro) of increased potassium currents and reduced cytosolic
calcium concentrations. However, Ishida et al. (1997) had
reported that when applied to hippocampal slices recom-
Fig. 1 APP processing and cleavage products. The nonamyloido- binant sAPPa enhanced LTP and shifted the frequency
genic APP processing pathway (right) involves cleavages by a- and
dependence for induction of long-term depression (LTD),
c-secretases resulting in the generation of a long secreted form of APP
(sAPPa) and C-terminal fragments (CTF 83, p3, and AICD50). The apparently by increasing cyclic GMP production. sAPPa
amyloidogenic APP processing pathway (left) involves cleavages by also has been shown to act as a growth factor for cells of
b- and c-secretases resulting in the generation of a long secreted epidermal origin (Herzog et al. 2004; Siemes et al. 2006) to
form of APP (sAPPb), C-terminal fragments (CTF 99 and CTF 89)
induce increased proliferation rates of embryonic and adult
and Abs. Ab fragments oligomerize and fibrillize leading to AD
pathology (left and upper panel). ex extracellular, PM plasma neural stem cells (Ohsawa et al. 1999; Caillé et al. 2004) as
membrane, cyt cytosol, DR6 death receptor 6 well as epidermal basal cells (Hoffmann et al. 2000) and
increased migration rates in keratinocytes (Kirfel et al.
considered, offering additional strategies for an Ab-oriented 2002). sAPPa is able to reverse most of the deficits seen in
therapy. However, over the recent years, more functions of APP knockout mice (Ring et al. 2007). We conclude that
the other APP cleavage products have emerged as well as sAPPa likely has growth promoting functions in dividing
more substrates of APP-cleaving enzymes. It must be rec- cells of epithelial origin, including embryonic and adult
ognized, therefore, that any intervention in APP processing neural stem cells and plays a role in brain development. In
is likely to affect more than just Ab production. In this differentiated neurons, sAPPa may have pro-survival
review, we will have a look at the current state of knowledge functions, enhance neurite outgrowth, and improve mem-
of APP processing enzyme activities and the functions of ory formation, although the exact mechanisms for these
various APP fragments. Interesting new findings have actions are not yet fully understood.
emerged recently, and it is, therefore, likely that more While sAPPa concentrations in cerebrospinal fluid
functions of APP fragments and APP processing enzymes (CSF) are reduced in carriers of the Swedish APP mutation
will be discovered in the near future. (APP 670/671) (Lannfelt et al. 1995), in the cases of spo-
radic AD, they seem to be unaltered (Sennvik et al. 2000;
Olsson et al. 2003) or increased (Lewczuk et al. 2008). The
sAPPa Dutch and Flemish mutations in the APP gene (APP
E693Q and APP A692G, respectively) are likely to (neg-
sAPPa is generated when a-secretase cleaves APP, a pro- atively) affect the a-secretase cleavage site. Interestingly,
cess also called shedding. The physiologic functions of families with these mutations display a somewhat different
sAPPa are poorly understood, but its actions have been clinical picture, with congophilic amyloid angiopathy and
Neuromol Med (2010) 12:1–12 3

in the case of the Flemish mutations also AD-like dementia that in a context of increased Ab production, reducing
and neuropathology. In these cases, both the altered Ab sAPPa (and possibly further increasing Ab) is the dominant
sequence and reduced a-secretase cleavage of APP may biologic effect of dominant negative a-secretase, whereas
contribute to the clinical picture. other effects of a-secretase (i.e., cleavage of proteins other
than APP) become dominant, when the dominant negative
mutant is expressed in a wild-type context, or when
a-Secretase a-secretase is overexpressed. If this interpretation is correct,
then it has to be assumed that one or more of a-secretase’s
a-Secretase activity is generally attributed to the ADAM cleavage products have the potential to facilitate seizures,
family of proteases. Among this family, ADAM 9, 10, 17, but Ab has a greater potential to do so.
and 19 have been shown to exert a-secretase activity
(Fahrenholz et al. 2000; Asai et al. 2003; Tanabe et al.
2006). At the moment, it is unclear which of these is the P3
relevant protease in AD patients. However, in a mouse
model of AD, Adam 10-overexpression has been shown to No clear biologic role has been established for the P3
reduce Ab production and plaque deposition in addition to fragment that is generated by a- and c-secretase cleavage of
lessening cognitive deficits (Postina et al. 2004). Therefore, APP.
ADAM 10, at the moment, is a good candidate to be a
relevant a-secretase in AD pathogenesis. However, given
the existing redundancy of ADAMs, it is quite possible that sAPPb
more than one ADAM is involved in sAPPa generation.
a-secretase activity has a constitutive and an inducible sAPPb is the secreted amino-terminal fragment that is
component that depends on its subcellular localization. generated when b-secretase cleaves APP. sAPPb lacks
a-secretase at the plasma membrane surface is considered most of the neuroprotective effects that have been associ-
constitutively active, and it is believed to be the main ated with sAPPa (Furukawa et al. 1996). A recent report
proteolytic pathway for APP that reaches the plasma demonstrated that sAPPb is critically involved in pruning
membrane (de Strooper and Annaert 2000). On the con- of synapses during development of both central and
trary, a-secretase that resides in the trans-Golgi network is peripheral neurons (Nikolaev et al. 2009). In this study,
regulated by PKC and competes with b-secretase in the sAPPb was found to act as a ligand for the death receptor 6
same location (Skovronsky et al. 2000). It is not clear (DR6) (a member of the TNFa family), which, upon
which specific ADAM protein is associated with constitu- binding, leads to cleavage and activation of caspase 6, but
tive and inducible a-secretase activity, but ADAM 10 can not caspase 3. This caspase 6 activation was associated
be activated by calcium (Le Gall et al. 2009). Neuronal with disintegration of axons, but not cell somata. Impor-
depolarization will lead to PKC activation and increased tantly, sAPPb generation was induced by trophic factor
vesicle and membrane turnover, and accordingly, neuronal withdrawal, and sAPPb could also bind to p75 neurotro-
activity was shown to increase a-secretase activity in an phin receptor (p75NTR), albeit with lower sensitivity and
NMDA receptor-dependent and calcium-sensitive fashion no clear biologic relevance in the context that was studied.
in mature neurons and neuron precursor cells (Gakhar- Other functions of sAPPb may include suppression of
Koppole et al. 2008; Hoey et al. 2009). However, the neuronal stem cell differentiation in favor of glial differ-
opposite (i.e., reduced a-secretase activity) upon neuronal entiation (Kwak et al. 2006), but these actions are not well
depolarization has been reported as well (Lesné et al. characterized.
2005). APP is only one of many substrates for ADAM
family proteins; others include N-cadherin (Kohutek et al.
2009), EGFR ligands, TNF-a, TGF-a, notch, and ephrin b-Secretase
(Edwards et al. 2008; Le Gall et al. 2009). While a dom-
inant negative mutant of ADAM 10 caused increased b-Secretase activity is mediated by BACE1 and cleaves
sensitivity to kainate-induced epileptic seizures in mice APP to release sAPPb and produce CTF99. BACE1 can
overexpressing the APP V717I (London) mutation, it also cleave APP at a more carboxy-terminal position,
reduced sensitivity to seizures in wild type mice. Similarly, resulting in CTF89 (and Ab 11–40 after c-secretase
surprising results were obtained with overexpression of cleavage) (Fig. 2). A related protein, BACE2, also can
wild type ADAM 10, which was associated with an exert b-secretase activity (Hussain et al. 2000), but it is
increased rate of seizures in both wild-type and APP V717I expressed at very low levels in the brain and is mostly
mice (Clement et al. 2008). One possible interpretation is confined to glial cells (Laird et al. 2005); interestingly,
4 Neuromol Med (2010) 12:1–12

APP posttranslational modifications occur and have an impact on


Extracellular tm Cytosol BACE1 activity (see Cole and Vassar 2008 and Hunt and
Turner 2009 for review). The subcellular localization of
BACE1 is predominantly within the trans-Golgi network and
endosomal compartment (Huse et al. 2002). Although
BACE1 reaches the plasma membrane due to vesicle traffic,
it is recycled quickly, and very little BACE1-mediated APP
…DAEFRHDSGYEVHHQKLVFFAEDVGSNKGAIIGLMVGGVVIATVIVITLVMLKKKQY… cleavage occurs at the plasma membrane; instead, APP is
β-site β’-site α-site γ- site ε-site cleaved by BACE primarily in endocytic vesicles. Interest-
ingly, within the secretory pathways of the endoplasmic
BACE1 ADAM9/10/17/19 γ-secretase
reticulum and trans-Golgi network, APP and BACE1 are
Fig. 2 APP processing enzymes and their cleavage sites. The amino located in separate membrane microdomains, due to APP
acid sequence of Ab and the carboxy-terminal adjacent region are binding to X11/Munc18 proteins (Saito 2008). Neuronal
displayed in single letter amino acid code. Left lines indicate the b and depolarization leads to Munc18 phosphorylation and causes
b’ cleavage site of BACE1. The middle line indicates the cleavage
site for a-secretase, which belongs to the ADAM family of proteases. APP to relocalize in BACE1-containing membrane micro-
Right lines indicate cleavage site for c-secretase, which is located domains (Sakurai 2008). Accordingly, neuronal depolar-
within the transmembrane region, as indicated by the black lines ization and vesicle turnover have been shown to increase
BACE1-mediated APP cleavage and Ab production
(Kamenetz et al. 2003). Thus, increased neuronal depolar-
BACE2 is a better a- than b-secretase (Farzan et al., 2000). ization increases APP cleavage by both a-secretase and
BACE2 appears irrelevant to AD pathogenesis and will not BACE1 in a competitive fashion (Kamenetz et al. 2003).
be considered in this review. Genetic ablation of BACE1 It will, therefore, be of interest to determine whether neu-
completely prevents the amyloid pathology in a mouse ronal activity can be fine-tuned to increase one activity over
model expressing the Swedish mutation of APP (APP670/ the other (Hoey et al. 2009). BACE1 is located in cholesterol-
671), together with mutated PS1 (Farzan et al. 2000; Laird rich lipid rafts (Abad-Rodriguez et al. 2004), but whether
et al. 2005), or in mice expressing only the Swedish APP lowering brain cholesterol levels in AD patients will actually
mutation (Tg2576) (Ohno et al. 2004). BACE1-deficient reduce Ab generation and improve cognitive function
mice lack b-secretase activity and Ab formation in neurons remains to be seen (Abad-Rodriguez et al. 2004; Cheng et al.
(Cai et al. 2001; Roberds et al. 2001; Luo et al. 2001. Also, 2007; Vetrivel et al. 2009).
BACE1 expression and activity are increased in brains of BACE1 has many other substrates apart from APP. They
AD patients (Holsinger et al. 2002; Yang et al. 2003).These include APP-like proteins 1 and 2 (APLP1 and 2), voltage-
studies provide the rationale for the current view that gated sodium channel b2-subunit, low density lipoprotein
BACE1 is the physiologically relevant b-secretase in the receptor-related protein, neuregulin 1, P-selectin glyco-
brain that contributes to AD pathology. At the end of this protein ligand-1, and sialyltransferase ST6Gal I (Hunt and
section, however, we will also have a look at cathepsins as Turner 2009). BACE1 knockout (BACE1-/-) mice show
enzymes with potential b-secretase activity. some interesting phenotypes. They show a rather severe
BACE1 expression is increased in situations of cellular hypomyelination of peripheral nerves (Willem et al. 2006),
stress, e.g., energy deprivation, hypoxia, and ischemia but only modest hypomyelination in the CNS (Hu et al.
(O’Connor et al. 2008; Guglielmotto et al. 2009), and 2006). They also have an increased rate of seizures, which
oxidative stress has been shown to increase BACE1 is even higher when BACE1-/- mice are crossed with
expression in a c-secretase dependent fashion (Jo et al. 2008). PDAPP mice (Kobayashi et al. 2008). BACE1 genetic
BACE1 is synthesized as a proenzyme in the endoplasmic ablation also causes a reduction in spine density in hip-
reticulum (that nonetheless possesses b-secretase activity). pocampal pyramidal neurons and alterations in behavior
Homodimerization, cleavage of the prodomain in the trans- tests that are believed to reveal schizophrenia-like pheno-
Golgi network by furin or furin-like proteases and complex types, including reduced pre-pulse inhibition, novelty-
glycosylation are further steps toward the fully active induced hyperactivity, hypersensitivity to the NMDA
enzyme (Westmeyer et al. 2004; Cole and Vassar 2008; Hunt receptor antagonist MK-801, as well as impairments in
and Turner 2009). BACE1 can be further cleaved to be cognition and social recognition (Savonenko et al. 2008). It
released from the plasma membrane (shedding). Shedding of is possible that these effects are mediated through impaired
BACE1 likely depends on proteases of the ADAM family processing of non-APP substrates, with neuregulin 1 being
which appears to be an interesting connection between a very likely candidate. It is interesting to note, however,
a-secretase and BACE1. The function and relevance of that the (modest) deficit that BACE1-/- mice display in the
BACE1 shedding remain unclear at the moment. Other Morris water-maze test was not seen when BACE1-/- mice
Neuromol Med (2010) 12:1–12 5

were crossed with APPswe/PS1DE9 transgenic mice, while The resulting peptide is predominantly 40 amino acids long
the deficits in social recognition remained unaltered (Laird but peptide length can range from 38 to 43 amino acids.
et al. 2005). These leaves open the possibility that Certain forms (e.g., Ab 1–42, 3–40) are considered more
increased APP expression and/or processing through a- and amyloidogenic than others (e.g., Ab1–40 or 1–38). Since
c-secretase can compensate for some of the deficits of both BACE1 and c-secretase activity are located in the
BACE1 deficiency. endosomal compartment and trans-Golgi network, it is
The view that BACE1 is the only relevant b-secretase in believed that most Ab is generated in these subcellular
AD has been challenged recently as other proteases, spe- localizations and subsequently secreted through exocytosis.
cifically cathepsins, have been shown to exert b-secretase Intracellular Ab has been observed as well, both in animal
activity (Hook et al. 2005, 2007a, b, 2008; Schechter and models of AD and human patients (Wirths et al. 2001;
Ziv 2008; Böhme et al. 2008; Klein et al. 2009). A possible Gómez-Ramos and Asunción Morán 2007), the signifi-
explanation for these findings and the seemingly discrepant cance of which remains rather uncertain (Wegiel et al.
results of Cai et al. (2001), Roberds et al. (2001), and Luo 2007). Ab can be cleaved by proteases such as insulin
et al. (2001) is that cathepsins cleave wildtype (wt) human degrading enzyme (Kurochkin and Goto 1994), neprilysin
APP but not human APP that is mutated at the b-secretase (Iwata et al. 2000), BACE1 (Huse et al. 2002) and
cleavage site (mtAPP) as is the case in transgenic AD cathepsin B (only Ab 1–42) (Mueller-Steiner et al. 2006).
models that use an APP form containing the Swedish While insulin degrading enzyme, neprilysin, and cathepsin
mutation (K670 N/M671L). Accordingly, genetic ablation B render Ab non-amyloidogenic, the significance of Ab
of cathepsin B has been reported to reduce Ab 1–40 and 11–40, which is generated by BACE1 cleavage of Ab at the
Ab 1–42 levels in the brains of mice that overexpress b’ site, has not yet been established. Other modifications
human wtAPP but not mtAPP with the Swedish and Indi- include pyroglutamate formation of the amino-terminal
ana (V717F at the c-secretase cleavage site) mutation glutamic acid residue through glutaminyl cyclase resulting
(Hook et al. 2009). The finding that the overexpression of in amino-terminal truncated pyroglutmate Ab 3–40/42,
BACE1 does not result in increased levels of endogenous which is highly amyloidogenic; inhibition of glutaminyl
(murine) Ab may point to a similar direction (Hirata-Fukae cyclase improves amyloidosis and cognition in mouse
et al. 2008). Also, the conversion of the aspartate residue at models of AD (Schilling et al. 2008). Conversion of
the b-secretase cleavage site to isoaspartate by a nonen- aspartate to isoaspartate at residue 23 in the Ab sequence
zymatic reaction has been reported to be a frequent event also has been reported to increase Ab aggregation (Shimizu
resulting in loss of BACE1 but not cathepsin B activity at et al. 2002). Ab production is either increased or the ratio
the b-secretase site (Böhme et al. 2008). However, the of Ab42–40 is increased in most hereditary cases and
picture becomes more complex if it is considered that animal models of AD. In AD patients, Ab levels in the CSF
cathepsin B can also degrade Ab 1–42 and genetic ablation are not increased, rather Ab42 reduction in CSF is a good
of cathepsin B has been reported to lead to increased pla- biomarker for AD (Shaw et al. 2009). This reduction in
que load and Ab 1–42 levels in mice with the Swedish and Ab42 likely reflects reduced clearance of Ab through the
Indiana mutation of APP (Mueller-Steiner et al. 2006). CSF and increased accumulation in amyloid plaques.
Accordingly, genetic ablation of the cathepsin B inhibitor The currently predominant hypothesis states that Ab
cystatin C leads to reduced plaque load and Ab 1–42 levels forms soluble oligomers in the extracellular space and that
in the same mice (Sun et al. 2008). Thus, the role of these oligomers inhibit NMDA-mediated synaptic trans-
cathepsin B in AD needs to be further investigated as it mission and ultimately cause spine and synapse loss
may contribute to both Ab generation and degradation. The through mechanisms not yet fully understood (Selkoe
seemingly conflicting results of some of these studies are a 2000). In a study using extracts from brains of AD patients
reminder that mouse models only imperfectly replicate the and hippocampal slice cultures, Ab dimers were shown to
human disease, and it is important to realize that unlike all be the most potent form of Ab oligomers to inhibit NMDA-
mouse models, the majority of sporadic AD cases neither mediated synaptic transmission, and higher molecular
contain mutations in the APP nor Presenilin genes. It is, weight oligomers and insoluble aggregates were able to
therefore, important to further investigate potential differ- release Ab dimers (Shankar et al. 2008). Most studies of
ences in wtAPP versus mtAPP processing. Ab toxicity have been performed with synthetic peptides of
various lengths (e.g., 1–40, 1–42, and 25–35). In general,
caution must be exercised when translating such results to
Ab in vivo situations as oligomerization status and post
translational modifications of endogenous Ab are different
Ab is at the center of AD pathogenesis. It is generated than synthetic peptides. Notably, concentrations required to
through sequential cleavage of APP by b- and c-secretases. elicit Ab toxicity in vitro with synthetic peptides are in the
6 Neuromol Med (2010) 12:1–12

5–10 lM range, which is higher than Ab concentrations includes binding to the protein Fe65, then recruitment of
likely encountered by neurons in vivo. Also, in human the histone deaceytelase TIP60, nuclear translocation and
primary neuron cultures 10 lM Ab 1–38 and 25–35 are transcriptional activation of target genes such as p53,
alone not toxic, but a preincubation for 3 days leads to GSK3b, neprilysin, EGFR, and others. Apart from this
increased sensitivity to glutamate toxicity (Mattson et al., pathway many other AICD binding proteins and effects
1992). One interesting interaction that has been reported have been described. However, these pathways have been
recently is between Ab and cellular prion protein (PrP) studied mainly in cell lines, sometimes in primary neurons,
(Laurén et al. 2009). In this study, Ab oligomers of *100 but hardly in vivo in the brain, making any extrapolation of
molecules would exert their inhibitory effect on NMDA- existing results into the context of AD brains very specu-
mediated synaptic transmission only when they could bind lative. Moreover, not all research groups can confirm even
to the cellular form of PrP. In PrP deficient mice, this the prototypical pathway of AICD binding to Fe65 and
interaction was absent, and Ab peptides were not inhibitory nuclear translocation with target gene transcription (Wal-
or toxic. Other interactions between APP or Ab and PrP, dron et al. 2008), leaving a fair amount of controversy in
and reciprocal modulation of AD or scrapie disease pro- the debate. Transgenic mice that overexpressed both AICD
gression in mouse have been described as well (Baier et al. and Fe65 have abnormal EEG recordings and increased
2008, Tamgüney et al. 2008); moreover, cellular PrP was susceptibility for seizures, features also seen in mice
shown to inhibit BACE1-mediated Ab production (Parkin overexpressing APP with the Swedish mutation but not in
et al. 2007). mice with the Swedish and Indiana mutation as well as a
disruption of the caspase cleavage site in the AICD
sequence (D664A) (Vogt et al. 2009). Overexpressing
CTF83 and CTF99 AICD with Fe65 also leads to hyperphosphorylation of the
microtubule associated protein tau and to neuronal cell loss
No biologically relevant roles are currently established for in animals 18 months and older (Ghosal et al. 2009).
the carboxy-terminal fragments CTF83 and CTF99 gener- Importantly transgenic mice overexpressing only Fe65 did
ated by a- and b-secretase, respectively. A more carboxy- not show any of these features. However, since no data are
terminal cleavage of APP by b-secretase cleavage results in available on the effect of AICD overexpression only we
a CTF89, the function of which also is unclear. feel that, as of now, the role of AICD in AD pathogenesis
remains to be firmly established, and future experiments
using human APP constructs with mutations in the
AICD e-cleavage site or the YENPTY domain should provide a
clearer picture. In an elegant approach, wtAPP lacking the
AICD is generated when c-secretase cleaves CTF83 or YENPTY domain was reintroduced into APP knockout
CTF99. Although c-secretase cleavage of APP to release mice (Ring et al. 2007). As a result, Ab generation was
Ab results in an AICD fragment of 57 or 59 amino acids, quite drastically reduced compared with wild-type mice,
another cleavage site (e-cleavage site), just a few amino probably due to the increased cell surface expression of
acids toward the carboxy-terminal end, will result in a 50- APP and reduced entry into the endocytic pathway,
amino acid AICD fragment (Fig. 2). This is mostly con- pointing to a role for AICD in APP trafficking. The results
sidered the typical AICD, but more cleavage sites for indicate that a clear-cut separate role for AICD may be
c-secretase and also for caspase 3 have been reported, difficult to establish as the domain may serve multiple roles
resulting in even shorter peptides (e.g., AICD 31 after in full-length APP and after cleavage. This consideration
caspase 3 cleavage). Regardless of the cleavage site, AICD applies, of course, not just to AICD but to other products of
of various lengths still contains the consensus motif YE- APP cleavage as well.
NPTY that is thought to be crucial for AICD binding to
adapter proteins such as Fe65 and subsequent biologic
actions. The term AICD has been coined by analogy to c-Secretase
protease cleavage of the protein Notch, another c-secretase
substrate, which results in release of the Notch intracellular c-secretase cleaves APP in its intra-membrane region at the
domain (NICD). c-secretase-mediated cleavage of Notch is c-cleavage site to generate Ab 1-40/42 or p3 and AICD59/
a critical step in the Notch signaling cascade, and this 57, a second cleavage at the e-cleavage site results in
similarity has fostered much research into AICD. The AICD50 (Fig. 2). At least four proteins need to interact for
AICD peptide can undergo more posttranslational modifi- c-secretase activity to unfold: PS1 or 2, Nct, Aph-1, and
cations, including phosphorylation (see Müller et al. 2008 Pen2 (Wolfe 2008). Aph-1 exists in two isoforms, Aph-1a
for review). The prototypical signaling pathway for AICD and b, encoded by two genes in humans; mice have a third
Neuromol Med (2010) 12:1–12 7

isoform, Aph-1c. PS1 and 2 have the actual protease memory and learning in behavior tests was statistically
activity, and in the brain, PS1 is the dominant presenilin. significant only in young (3 months) mice, while in older
The full-length PS proteins contain nine transmembrane mice (15–17 months) the effect was reduced and not sta-
regions and are cleaved in the cytoplasmic loop between tistically significant (Saura et al. 2005). Another recent
the 6th and 7th transmembrane region to generate an report indicates that genetic ablation of Aph-1b and c in
amino-terminal and carboxy-terminal fragments (Thinak- mice reduces c-secretase-mediated APP cleavage (specifi-
aran et al. 1996). Both fragments are stable and stay in cally the generation of the more amyloidogenic Ab1–42),
close association in the membrane and represent the active but does not affect Notch cleavage (Serneels et al. 2009).
form of PS, whereas full-length PS is rapidly degraded. In Genetic ablation of Aph-1b/c resulted in subtle behavioral
the current model of c-secretase activity, both the amino- abnormalities that were reminiscent of those seen in
terminal and carboxy-terminal fragments of PS are BACE1-/- mice (Laird et al. 2005, Savonenko et al. 2008)
required for the actual aspartyl protease activity, whereas and may reflect a deficit in processing of neuregulin (which
Nct and Aph-1 are attributed a substrate recognition is also a BACE1 substrate) or its binding partner ErbB4.
function. Fully active c-secretase matures in discrete steps: However, Aph-1b/c knockout resulted in a complete
first Nct and Aph-1 form a complex, then PS binds to this reversal of cognitive deficits in APP/PS1 transgenic mice,
complex, and finally Pen2 will complete the complex and together with a reduction in amyloid pathology. The oldest
may facilitate auto-cleavage of PS. (Li et al. 2009). More mice used in this study were 11 months old, and at the
proteins interact with c-secretase and may modulate its moment, no data are published on whether the beneficial
function (Winkler et al. 2009). More than 50 c-secretase effects of Aph-1b/c knockout can be sustained in older
substrates have been discovered (Beel and Sanders 2008; APP/PS1 transgenic animals.
Wakabayashi and de Strooper 2008), including APP, The orphan G-protein coupled receptor 3 (Gpr3) was
Notch, the neuregulin binding partner ErbB4, N-cadherin, recently shown to affect c-secretase assembly and subcel-
p75NTR, and others. In a study using different knockouts lular location (Thathiah et al. 2009). Gpr3, although not an
of c-secretase components heterozygous deletion of PS1, essential part of the c-secretase complex, facilitated the
Aph-1a or Nct resulted in a reduction of c-secretase activity assembly of the four required proteins to the fully active
ranging from 25% (PS1?/-) to 64% for PS1?/-; Nct?/- protein complex and targeted c-secretase preferentially to
mice (Li et al. 2009). In this study, a c-secretase reduction lipid rafts and the plasma membrane leading to increased
of 30% or lower (PS1?/- or Aph-1a?/- mice) was not Ab 1–40 and 1–42 production in vitro and in vivo, whereas
associated with an increased rate of squamous cell carci- genetic ablation of Gpr3 led to a reduction of Ab pro-
nomas (that is caused by deficient Notch signaling), while duction in young mice. Notch signaling was not affected by
Nct?/- and PS1?/-; Nct?/- mice with 50 and 64% Gpr3 knockout. The effects of Gpr3 expression on cogni-
reductions in c-secretase activity, respectively, did have tion and behavior have not been reported yet.
increased carcinogenesis. Interestingly, a moderate reduc- Taken together, good evidence has now been provided
tion of c-secretase activity by 30% through Aph-1a het- that c-secretase activity exists as separate subspecies with
erozygous knockout was enough to reduce amyloid different subunit composition and, thus, different struc-
pathology in APPswe/PS1DE9 mice. A series of studies tures, substrate specificities, and binding partners. This
used a conditional knockout approach of c-secretase pro- provides a good rationale for the development of c-secre-
teins using the Cre recombinase system under a promoter tase inhibitors with relative specificity for APP over Notch,
that is expressed only in the postnatal forebrain (Yu et al. and compounds with such a profile have been described
2001; Saura et al. 2005; Tabuchi et al. 2009). From these already (Yang et al. 2008; Cole et al. 2009). However,
studies, it can be concluded that a complete inactivation of results from the conditional knockout studies suggest that
c-secretase through conditional knockout of Nct (Tabuchi reduced c-secretase activity is associated with detrimental
et al. 2009) in a normally developed brain leads to pro- side effects in the brain especially in older animals. It
gressive neurodegeneration. Conditional knockout of PS1 remains to be seen whether these effects reflect a deficit in
does not lead to a progressive neurodegenerative pheno- APP cleavage and processing or are a due to reduced
type, possibly because the loss of PS1 can be partially cleavage of other substrates (e.g., neuregulin or ErbB4) and
compensated for by PS2. Nevertheless, these mice exhib- how the human brain is affected by c-secretase inhibition.
ited some cognitive deficits in behavior testing (Yu et al. PS1 also has a role in endoplasmic reticulum-mediated
2001). Importantly, these deficits were unlikely to reflect a calcium release. At the moment, there is still some con-
deficit in Notch signaling, as expression of Notch target troversy about the exact mechanism, as PS1 may act as a
genes was unaffected. When the conditional PS1 knockout calcium leak channel or it may enhance the opening of the
mice were crossed with APP transgenic mice, Ab pro- inositol-3-phosphate receptor (IP3R); however, it does
duction was clearly reduced, but the beneficial effect on seem clear that PS1 causes an increase in cytosolic calcium
8 Neuromol Med (2010) 12:1–12

concentrations (Tu et al. 2006, Nelson et al. 2007, Cheung 2008). TAG1 binding to APP induced APP processing
et al. 2008). Interestingly, although this function has been (preferentially through a- and c-secretase), and through
reported to be independent of c-secretase activity, it is also genetic manipulations it was shown that the AICD-medi-
affected by AD-associated PS1 mutations which cause ated signaling through Fe65 inhibited neuronal differenti-
higher cytosolic calcium concentrations when cells are ation of embryonic stem cells and kept the stem cells in a
stimulated (Guo et al. 1997; Chan et al. 2000). While c- mode of self-renewal.
secretase activity is not required for PS1-mediated gating
of (IP3Rs), IP3Rs gating through PS1 is important for
c-secretase function (Cheung et al. 2008). These data provide Conclusion
a further link between disturbed calcium homeostasis and
AD, where increased cytosolic calcium concentrations may Great progress has been made in characterizing the mole-
be associated with increased sensitivity to excitotoxicity, cules involved in APP processing and the functions of APP
epileptiform discharges and seizures as well as skewed cleavage products. As a result, a complex picture has
information processing, as relatively smaller amounts of emerged for the physiologic and pathologic roles of APP.
synaptic input are required to elicit an action potential or A clearly defined role for sAPPb in axon pruning has been
calcium dependent signals, e.g., through calcium/calmodu- demonstrated, and good evidence exists for sAPPa having
lin dependent kinases. important roles in brain development and synaptic plas-
ticity, as well as growth factor-like properties. Whether
AICD acts as a transcriptional activator remains contro-
Full-Length APP versial. Recent data indicate that it may play a role in
embryonic neural stem cell development, but also regulate
The search for a physiologic role of full-length APP is APP trafficking and, thus, modulate APP processing.
inseparably linked to the functions of APP cleavage Neuronal activity modulates APP processing pathways,
products. Overexpression of human APP (either wt or with suggesting important roles for activity-induced APP
the Swedish mutation) in mice is associated with increased cleavage products in regulating neuronal excitability and
size of cortical neurons, whereas overexpression of PS1 is synaptic plasticity. The finding that cathepsin B can act as a
not (Oh et al. 2009). At the moment, it is not clear whether b-secretase for wtAPP, but not mtAPP, may have great
this is an effect of full-length APP or one of its cleavage impact on AD research and, therefore, merits further
products. Interestingly, increased size of neuron somata, investigation and corroboration. Ab remains the main
nuclei and nucleoli in the hippocampus and cortex has also suspect in AD pathogenesis, and the molecular mecha-
been observed in postmortem brains of humans that show nisms and binding partners of soluble Ab toxicity are
AD-like pathology but had no cognitive deficits (Riudavets beginning to be unraveled. Posttranslational modifications
et al. 2007, Iacono et al. 2008, Iacono et al. 2009). of Ab may contribute to Ab oligomerization and toxicity,
APP deficient mice show a number of phenotypes, as may the extent to which Ab is cleared by Ab-degrading
including reduced body and brain seize, impaired learning proteases. With regard to APP processing enzymes, it has
and LTP, reduced grip strength, hypersensitivity to sei- become very clear that all involved enzymes have multiple
zures, and increased frequency of corpus callosum dys- substrates and are not specific for APP, and that PS1 has
genesis (Ring et al. 2007). These phenotypes mostly functions beyond c-secretase. While initially concerns were
become apparent during early postnatal development, great that c-secretase inhibitors inevitably would impair
rather than in adult stages. By reintroducing only the Notch processing, a better understanding of c-secretase
sAPPa fragment into APP deficient mice, it was demon- mechanics has helped to make the development of Notch
strated that lack of sAPPa is responsible for most of the sparing c-secretase inhibitors seem a very feasible task.
phenotypes observed in APP mice (Ring et al. 2007). However, while Notch appears to become less of a problem
While single genetic ablation of APP or APP-like protein to drug development, the sheer quantity of substrates for
(APLP) 1 and 2 results in viable mice, deletion of APP and both b- and c-secretase is a reason for concern. Combining
APLP2, APLP1 and APLP2, or APP and APLPs 1 and 2 moderate b- and c-secretase inhibition may be a tangible
results in perinatal lethality suggesting redundancy in some strategy to reduce potential side effects of secretase inhi-
of the physiologic functions of these proteins. Therefore, it bition while additively reducing Ab production. However,
seems that there is an important role for APP in brain since c-secretase requires a substrate to be cleaved first by a
development that is in large part mediated by sAPPa and sheddase, b- and c-secretase may, in fact, share multiple
partly overlaps with functions of APLPs 1 and 2. Transient substrates, neuregulin/ErbB4 and the voltage-gated sodium
axonal glycoprotein l (TAG1) was recently discovered to channel b2-subunit being good examples. A detailed
act as a binding partner for full-length APP (Ma et al. understanding of the molecular aspects of secretase activity
Neuromol Med (2010) 12:1–12 9

may help to generate secretase inhibitors with high speci- Alzheimer’s disease by presenilin regulation of InsP3 receptor
ficity for APP. Also, it is important to understand the channel gating. Neuron, 58, 871–883.
Clement, A. B., Hanstein, R., Schröder, A., Nagel, H., Endres, K.,
physiologic functions of secretases in the aged human brain Fahrenholz, F., et al. (2008). Effects of neuron-specific
to judge the potential for mechanism-based side effects of ADAM10 modulation in an in vivo model of acute excitotoxic
secretase inhibitors. The major challenge of course remains stress. Neuroscience, 152, 459–468.
treating AD patients, who at the time of diagnosis are in an Cole, D. C., Stock, J. R., Kreft, A. F., Antane, M., Aschmies, S. H.,
Atchison, K. P., et al. (2009). (S)-N-(5-Chlorothiophene-2-
advanced disease stage when compared to AD mouse sulfonyl)-beta, beta-diethylalaninol a Notch-1-sparing gamma-
models. Therefore, improved diagnosis criteria and tools secretase inhibitor. Bioorganic and Medicinal Chemistry Letters,
seem indispensable requirements for better efficacy of any 19, 926–929.
anti-amyloid strategy. Considering all challenges that have Cole, S. L., & Vassar, R. (2008). The role of amyloid precursor
protein processing by BACE1, the beta-secretase, in Alzheimer
to be met by anti-amyloid or secretase-focused therapies, disease pathophysiology. The Journal of Biological Chemistry,
the development of non-amyloid-based therapeutic strate- 283, 29621–29625.
gies would be a very welcome addition to the portfolio of De Strooper, B., & Annaert, W. (2000). Proteolytic processing and
AD research. cell biological functions of the amyloid precursor protein.
Journal of Cell Science, 113, 1857–1870.
Edwards, D. R., Handsley, M. M., & Pennington, C. J. (2008). The
Acknowledgment This study was in part supported by the Intra- ADAM metalloproteinases. Molecular Aspects of Medicine, 29,
mural Research Program of the NIH, National Institute on Aging. 258–289.
Fahrenholz, F., Gilbert, S., Kojro, E., Lammich, S., & Postina, R.
(2000). Alpha-secretase activity of the disintegrin metallopro-
tease ADAM 10. Influences of domain structure. Annals of the
References New York Academy of Sciences, 920, 215–222.
Farzan, M., Schnitzler, C. E., Vasilieva, N., Leung, D., & Choe, H.
Abad-Rodriguez, J., Ledesma, M. D., Craessaerts, K., Perga, S., (2000). BACE2, a b-secretase homolog, cleaves at the b site and
Medina, M., Delacourte, A., et al. (2004). Neuronal membrane within the amyloid-b region of the amyloid-b precursor protein.
cholesterol loss enhances amyloid peptide generation. Journal of Proceedings of the National Academy of Sciences of the United
Cell Biology, 167, 953–960. States of America, 97, 9712–9717.
Asai, M., Hattori, C., Szabó, B., Sasagawa, N., Maruyama, K., Furukawa, K., Barger, S. W., Blalock, E. M., & Mattson, M. P.
Tanuma, S., et al. (2003). Putative function of ADAM9, (1996). Activation of K? channels and suppression of neuronal
ADAM10, and ADAM17 as APP alpha-secretase. Biochemical activity by secreted beta-amyloid-precursor protein. Nature, 379,
and Biophysical Research Communications, 301, 231–235. 74–78.
Baier, M., Apelt, J., Riemer, C., Gültner, S., Schwarz, A., Bamme, T., Gakhar-Koppole, N., Hundeshagen, P., Mandl, C., Weyer, S. W.,
et al. (2008). Prion infection of mice transgenic for human Allinquant, B., Müller, U., et al. (2008). Activity requires soluble
APPSwe: Increased accumulation of cortical formic acid amyloid precursor protein alpha to promote neurite outgrowth in
extractable Abeta(1–42) and rapid scrapie disease development. neural stem cell-derived neurons via activation of the MAPK
International Journal of Developmental Neuroscience, 26, 821– pathway. European Journal of Neuroscience, 28, 871–882.
824. Ghosal, K., Vogt, D. L., Liang, M., Shen, Y., Lamb, B. T. &
Beel, A. J., & Sanders, C. R. (2008). Substrate specificity of gamma- Pimplikar, S. W. (2009). Alzheimer’s disease-like pathological
secretase and other intramembrane proteases. Cellular and features in transgenic mice expressing the APP intracellular
Molecular Life Sciences, 65, 1134–1311. domain. Proceedings of the National Academy of Sciences of the
Böhme, L., Hoffmann, T., Manhart, S., Wolf, R., & Demuth, H. U. United States of America (in press).
(2008). Isoaspartate-containing amyloid precursor protein- Gómez-Ramos, P., & Asunción Morán, M. (2007). Ultrastructural
derived peptides alter efficacy and specificity of potential beta- localization of intraneuronal Abeta-peptide in Alzheimer disease
secretases. Biological chemistry, 389, 1055–1066. brains. Journal of Alzheimer’s Disease, 11, 53–59.
Cai, H., Wang, Y., McCarthy, D., Wen, H., Borchelt, D. R., Price, D. Guglielmotto, M., Aragno, M., Autelli, R., Giliberto, L., Novo, E.,
L., et al. (2001). BACE1 is the major beta-secretase for Colombatto, S., et al. (2009). The up-regulation of BACE1
generation of Abeta peptides by neurons. Nature Neuroscience, mediated by hypoxia and ischemic injury: Role of oxidative
4, 233–234. stress and HIF1alpha. Journal of Neurochemistry, 108, 1045–
Caillé, I., Allinquant, B., Dupont, E., Bouillot, C., Langer, A., Müller, 1056.
U., et al. (2004). Soluble form of amyloid precursor protein Guo, Q., Sopher, B. L., Furukawa, K., Pham, D. G., Robinson, N.,
regulates proliferation of progenitors in the adult subventricular Martin, G. M., et al. (1997). Alzheimer’s presenilin mutation
zone. Development, 131, 2173–2181. sensitizes neural cells to apoptosis induced by trophic factor
Chan, S. L., Mayne, M., Holden, C. P., Geiger, J. D., & Mattson, M. withdrawal and amyloid beta-peptide: Involvement of calcium
P. (2000). Presenilin-1 mutations increase levels of ryanodine and oxyradicals. Journal of Neuroscience, 17, 4212–4222.
receptors and calcium release in PC12 cells and cortical neurons. Herzog, V., Kirfel, G., Siemes, C., & Schmitz, A. (2004). Biological
The Journal of Biological Chemistry, 275, 18195–18200. roles of APP in the epidermis. European Journal of Cell Biology,
Cheng, H., Vetrivel, K. S., Gong, P., Meckler, X., Parent, A., & 83, 613–624.
Thinakaran, G. (2007). Mechanisms of disease: new therapeutic Hirata-Fukae, C., Sidahmed, E. H., Gooskens, T. P., Aisen, P. S.,
strategies for Alzheimer’s disease–targeting APP processing in Dewachter, I., Devijver, H., et al. (2008). Beta-site amyloid
lipid rafts. Nature Clinical Practice. Neurology, 3, 374–382. precursor protein-cleaving enzyme-1 (BACE1)-mediated
Cheung, K. H., Shineman, D., Müller, M., Cárdenas, C., Mei, L., changes of endogenous amyloid beta in wild-type and transgenic
Yang, J., et al. (2008). Mechanism of Ca2? disruption in mice in vivo. Neuroscience Letters, 435, 186–189.
10 Neuromol Med (2010) 12:1–12

Hoey, S. E., Williams, R. J., & Perkinton, M. S. (2009). Synaptic leads to biochemical and pathological deposition. Nature
NMDA receptor activation stimulates alpha-secretase amyloid Medicine, 6, 143–150.
precursor protein processing and inhibits amyloid-beta produc- Jo, D. G., Arumugam, T. V., Woo, H. N., Park, J. S., Tang, S. C.,
tion. Journal of Neuroscience, 29, 4442–4460. Mughal, M., et al. (2008). Evidence that gamma-secretase
Hoffmann, J., Twiesselmann, C., Kummer, M. P., Romagnoli, P., & mediates oxidative stress-induced beta-secretase expression in
Herzog, V. (2000). A possible role for the Alzheimer amyloid Alzheimer’s disease. Neurobiology of Aging (in press).
precursor protein in the regulation of epidermal basal cell Kamenetz, F., Tomita, T., Hsieh, H., Seabrook, G., Borchelt, D.,
proliferation. European Journal of Cell Biology, 79, 905–914. Iwatsubo, T., et al. (2003). APP processing and synaptic
Holsinger, R. M., McLean, C. A., Beyreuther, K., Masters, C. L., & function. Neuron, 37, 925–937.
Evin, G. (2002). Increased expression of the amyloid precursor Kirfel, G., Borm, B., Rigort, A., & Herzog, V. (2002). The secretory
beta-secretase in Alzheimer’s disease. Annals of Neurology, 51, beta-amyloid precursor protein is a motogen for human epider-
783–786. mal keratinocytes. European Journal of Cell Biology, 81, 664–
Hook, G., Hook, V. Y., & Kindy, M. (2007a). Cysteine protease 676.
inhibitors reduce brain beta-amyloid and beta-secretase activity Klein, D. M., Felsenstein, K. M., & Brenneman, D. E. (2009).
in vivo and are potential Alzheimer’s disease therapeutics. Cathepsins B and L differentially regulate amyloid precursor
Biological Chemistry, 388, 979–983. protein processing. Journal of Pharmacology and Experimental
Hook, V., Kindy, M., & Hook, G. (2007b). Cysteine protease Therapeutics, 328, 813–821.
inhibitors effectively reduce in vivo levels of brain beta-amyloid Kobayashi, D., Zeller, M., Cole, T., Buttini, M., McConlogue, L.,
related to Alzheimer’s disease. Biological Chemistry, 388, 247– Sinha, S., et al. (2008). BACE1 gene deletion: impact on
252. behavioral function in a model of Alzheimer’s disease. Neuro-
Hook, V. Y., Kindy, M., & Hook, G. (2008). Inhibitors of cathepsin B biology of Aging, 29, 861–873.
improve memory and reduce beta-amyloid in transgenic Alzhei- Kohutek, Z. A., di Pierro, C. G., Redpath, G. T., & Hussaini, I. M.
mer disease mice expressing the wild-type, but not the Swedish (2009). ADAM-10-mediated N-cadherin cleavage is protein
mutant, beta-secretase site of the amyloid precursor protein. The kinase C-alpha dependent and promotes glioblastoma cell
Journal of Biological Chemistry, 283, 7745–7753. migration. The Journal of Neuroscience, 29, 4605–4615.
Hook, V. Y., Kindy, M., Reinheckel, T., Peters, C., & Hook, G. Kurochkin, I. V., & Goto, S. (1994). Alzheimer’s beta-amyloid
(2009). Genetic cathepsin B deficiency reduces beta-amyloid in peptide specifically interacts with and is degraded by insulin
transgenic mice expressing human wild-type amyloid precursor degrading enzyme. FEBS Letter, 345, 33–37.
protein. Biochemical and Biophysical Research Communica- Kwak, Y. D., Brannen, C. L., Qu, T., Kim, H. M., Dong, X., Soba, P.,
tions, 386, 284–288. et al. (2006). Amyloid precursor protein regulates differentiation
Hook, V., Toneff, T., Bogyo, M., Greenbaum, D., Medzihradszky, K. of human neural stem cells. Stem Cells and Development, 15,
F., Neveu, J., et al. (2005). Inhibition of cathepsin B reduces 381–389.
beta-amyloid production in regulated secretory vesicles of Laird, F. M., Cai, H., Savonenko, A. V., Farah, M. H., He, K.,
neuronal chromaffin cells: evidence for cathepsin B as a Melnikova, T., et al. (2005). BACE1, a major determinant of
candidate beta-secretase of Alzheimer’s disease. Biological selective vulnerability of the brain to amyloid-beta amyloido-
Chemistry, 386, 931–940. genesis, is essential for cognitive, emotional, and synaptic
Hu, X., Hicks, C. W., He, W., Wong, P., Macklin, W. B., Trapp, B. functions. Journal of Neuroscience, 25, 11693–11709.
D., et al. (2006). Bace1 modulates myelination in the central and Lannfelt, L., Basun, H., Wahlund, L. O., Rowe, B. A., & Wagner, S.
peripheral nervous system. Nature Neuroscience, 9, 1520–1525. L. (1995). Decreased alpha-secretase-cleaved amyloid precursor
Hunt, C. E., & Turner, A. J. (2009). Cell biology, regulation and protein as a diagnostic marker for Alzheimer’s disease. Nature
inhibition of beta-secretase (BACE-1). FEBS Journal, 276, Medicine, 1, 829–832.
1845–1859. Laurén, J., Gimbel, D. A., Nygaard, H. B., Gilbert, J. W., &
Huse, J. T., Liu, K., Pijak, D. S., Carlin, D., Lee, V. M., & Doms, R. Strittmatter, S. M. (2009). Cellular prion protein mediates
W. (2002). Beta-secretase processing in the trans-Golgi network impairment of synaptic plasticity by amyloid-beta oligomers.
preferentially generates truncated amyloid species that accumu- Nature, 457, 1128–1132.
late in Alzheimer’s disease brain. The Journal of Biological Le Gall, S. M., Bobé, P., Reiss, K., Horiuchi, K., Niu, X. D., Lundell,
Chemistry, 277, 16278–16284. D., et al. (2009). ADAMs 10 and 17 represent differentially
Hussain, I., Powell, D. J., Howlett, D. R., Chapman, G. A., Gilmour, regulated components of a general shedding machinery for
L., Murdock, P. R., et al. (2000). ASP1 (BACE2) cleaves the membrane proteins such as transforming growth factor alpha,
amyloid precursor protein at the b-secretase site. Molecular and L-selectin, and tumor necrosis factor alpha. Molecular Biology
Cellular Neurosciences, 16, 609–619. of the Cell, 20, 1785–1794.
Iacono, D., Markesbery, W. R., Gross, M., Pletnikova, O., Rudow, G., Lesné, S., Ali, C., Gabriel, C., Croci, N., MacKenzie, E. T., Glabe, C.
Zandi, P., et al. (2009). The nun study. Clinically silent AD, G., et al. (2005). NMDA receptor activation inhibits alpha-
neuronal hypertrophy, and linguistic skills in early life. Neurol- secretase and promotes neuronal amyloid-beta production.
ogy (in press). Journal of Neuroscience, 25, 9367–9377.
Iacono, D., O’Brien, R., Resnick, S. M., Zonderman, A. B., Lewczuk, P., Kornhuber, J., Vanderstichele, H., Vanmechelen, E.,
Pletnikova, O., Rudow, G., et al. (2008). Neuronal hypertrophy Esselmann, H., Bibl, M., et al. (2008). Multiplexed quantification
in asymptomatic Alzheimer disease. Journal of Neuropathology of dementia biomarkers in the CSF of patients with early
and Experimental Neurology, 67, 578–589. dementias and MCI: A multicenter study. Neurobiology of
Ishida, A., Furukawa, K., Keller, J. N., & Mattson, M. P. (1997). Aging, 29, 812–818.
Secreted form of beta-amyloid precursor protein shifts the Li, H., Wolfe, M. S., & Selkoe, D. J. (2009). Toward structural
frequency dependency for induction of LTD, and enhances LTP elucidation of the c-secretase complex. Structure, 17, 326–334.
in hippocampal slices. Neuroreport, 8, 2133–2137. Luo, Y., Bolon, B., Kahn, S., Bennett, B. D., Babu-Khan, S., Denis,
Iwata, N., Tsubuki, S., Takaki, Y., Watanabe, K., Sekiguchi, M., P., et al. (2001). Mice deficient in BACE1, the Alzheimer’s beta-
Hosoki, E., et al. (2000). Identification of the major Abeta1-42- secretase, have normal phenotype and abolished beta-amyloid
degrading catabolic pathway in brain parenchyma: Suppression generation. Nature Neuroscience, 4, 231–232.
Neuromol Med (2010) 12:1–12 11

Ma, Q. H., Futagawa, T., Yang, W. L., Jiang, X. D., Zeng, L., Takeda, Alzheimer disease mouse model. Journal of Clinical Investiga-
Y., et al. (2008). A TAG1-APP signalling pathway through Fe65 tion, 113, 1456–1464.
negatively modulates neurogenesis. Nature Cell Biology, 10, Ring, S., Weyer, S. W., Kilian, S. B., Waldron, E., Pietrzik, C. U.,
283–294. Filippov, M. A., et al. (2007). The secreted beta-amyloid
Mattson, M. P. (1997). Cellular actions of beta-amyloid precursor precursor protein ectodomain APPs alpha is sufficient to rescue
protein and its soluble and fibrillogenic derivatives. Physiolog- the anatomical, behavioral, and electrophysiological abnormal-
ical Reviews, 77, 1081–1132. ities of APP-deficient mice. Journal of Neuroscience, 27, 7817–
Mattson, M. P., Cheng, B., Culwell, A. R., Esch, F. S., Lieberburg, I., 7826.
& Rydel, R. E. (1993). Evidence for excitoprotective and Riudavets, M. A., Iacono, D., Resnick, S. M., O’Brien, R., Zonder-
intraneuronal calcium-regulating roles for secreted forms of the man, A. B., Martin, L. J., et al. (2007). Resistance to Alzheimer’s
beta-amyloid precursor protein. Neuron, 10, 243–254. pathology is associated with nuclear hypertrophy in neurons.
Mattson, M. P., Cheng, B., Davis, D., Bryant, K., Lieberburg, I., & Neurobiology of Aging, 28, 1484–1492.
Rydel, R. E. (1992). beta-Amyloid peptides destabilize calcium Roberds, S. L., Anderson, J., Basi, G., Bienkowski, M. J., Branstetter,
homeostasis and render human cortical neurons vulnerable to D. G., Chen, K. S., et al. (2001). BACE knockout mice are
excitotoxicity. Journal of Neuroscience, 12, 376–389. healthy despite lacking the primary beta-secretase activity in
Meziane, H., Dodart, J. C., Mathis, C., Little, S., Clemens, J., Paul, S. brain: Implications for Alzheimer’s disease therapeutics. Human
M., et al. (1998). Memory-enhancing effects of secreted forms of Molecular Genetics, 10, 1317–1324.
the beta-amyloid precursor protein in normal and amnestic mice. Saito, Y., Sano, Y., Vassar, R., Gandy, S., Nakaya, T., Yamamoto, T.,
Proceedings of the National Academy of Sciences of the United et al. (2008). X11 proteins regulate the translocation of amyloid
States of America, 95, 12683–12688. beta-protein precursor (APP) into detergent-resistant membrane
Mueller-Steiner, S., Zhou, Y., Arai, H., Roberson, E. D., Sun, B., and suppress the amyloidogenic cleavage of APP by beta-site-
Chen, J., et al. (2006). Antiamyloidogenic and neuroprotective cleaving enzyme in brain. The Journal of Biological Chemistry,
functions of cathepsin B: Implications for Alzheimer’s disease. 283, 35763–35771.
Neuron, 51, 703–714. Sakurai, T., Kaneko, K., Okuno, M., Wada, K., Kashiyama, T.,
Müller, T., Meyer, H. E., Egensperger, R., & Marcus, K. (2008). The Shimizu, H., et al. (2008). Membrane microdomain switching: A
amyloid precursor protein intracellular domain (AICD) as regulatory mechanism of amyloid precursor protein processing.
modulator of gene expression, apoptosis, and cytoskeletal Journal of Cell Biology, 183, 339–352.
dynamics-relevance for Alzheimer’s disease. Progress in Neu- Saura, C. A., Chen, G., Malkani, S., Choi, S. Y., Takahashi, R. H.,
robiology, 85, 393–406. Zhang, D., et al. (2005). Conditional inactivation of presenilin 1
Nelson, O., Tu, H., Lei, T., Bentahir, M., de Strooper, B., & prevents amyloid accumulation and temporarily rescues contex-
Bezprozvanny, I. (2007). Familial Alzheimer disease-linked tual and spatial working memory impairments in amyloid
mutations specifically disrupt Ca2? leak function of presenilin precursor protein transgenic mice. Journal of Neuroscience,
1. Journal of Clinical Investigation, 117, 1230–1239. 25, 6755–6764.
Nikolaev, A., McLaughlin, T., O’Leary, D. D., & Tessier-Lavigne, M. Savonenko, A. V., Melnikova, T., Laird, F. M., Stewart, K. A., Price,
(2009). APP binds DR6 to trigger axon pruning and neuron death D. L., & Wong, P. C. (2008). Alteration of BACE1-dependent
via distinct caspases. Nature, 457, 981–989. NRG1/ErbB4 signaling and schizophrenia-like phenotypes in
O’Connor, T., Sadleir, K. R., Maus, E., Velliquette, R. A., Zhao, J., BACE1-null mice. Proceedings of the National Academy of
Cole, S. L., et al. (2008). Phosphorylation of the translation Sciences of the United States of America, 105, 5585–5590.
initiation factor eIF2alpha increases BACE1 levels and promotes Schechter, I., & Ziv, E. (2008). Kinetic properties of cathepsin D and
amyloidogenesis. Neuron, 60, 988–1009. BACE 1 indicate the need to search for additional beta-secretase
Oh, E. S., Savonenko, A. V., King, J. F., Fangmark Tucker, S. M., candidate(s). Biological Chemistry, 389, 313–320.
Rudow, G. L., Xu, G., et al. (2009). Amyloid precursor protein Schilling, S., Zeitschel, U., Hoffmann, T., Heiser, U., Francke, M.,
increases cortical neuron size in transgenic mice. Neurobiology Kehlen, A., et al. (2008). Glutaminyl cyclase inhibition atten-
of Aging, 30, 1238–1244. uates pyroglutamate Abeta and Alzheimer’s disease-like pathol-
Ohno, M., Sametsky, E. A., Younkin, L. H., Oakley, H., Younkin, S. ogy. Nature Medicine, 14, 1106–1111.
G., Citron, M., et al. (2004). BACE1 deficiency rescues memory Selkoe, D. J. (2000). Toward a comprehensive theory for Alzheimer’s
deficits and cholinergic dysfunction in a mouse model of disease. Hypothesis: Alzheimer’s disease is caused by the
Alzheimer’s disease. Neuron, 41, 27–33. cerebral accumulation and cytotoxicity of amyloid beta-protein.
Ohsawa, I., Takamura, C., Morimoto, T., Ishiguro, M., & Kohsaka, S. Annals of the New York Academy of Sciences, 924, 17–25.
(1999). Amino-terminal region of secreted form of amyloid Sennvik, K., Fastbom, J., Blomberg, M., Wahlund, L. O., Winblad, B.,
precursor protein stimulates proliferation of neural stem cells. & Benedikz, E. (2000). Levels of alpha- and beta-secretase cleaved
European Journal of Neuroscience, 11, 1907–1913. amyloid precursor protein in the cerebrospinal fluid of Alzheimer’s
Olsson, A., Höglund, K., Sjögren, M., Andreasen, N., Minthon, L., disease patients. Neuroscience Letters, 278, 169–172.
Lannfelt, L., et al. (2003). Measurement of alpha- and beta- Serneels, L., Van Biervliet, J., Craessaerts, K., Dejaegere, T., Horré,
secretase cleaved amyloid precursor protein in cerebrospinal K., Van Houtvin, T., et al. (2009). gamma-Secretase heteroge-
fluid from Alzheimer patients. Experimental Neurology, 183, neity in the Aph-1 subunit: Relevance for Alzheimer’s disease.
74–80. Science, 324, 639–642.
Parkin, E. T., Watt, N. T., Hussain, I., Eckman, E. A., Eckman, C. B., Shankar, G. M., Li, S., Mehta, T. H., Garcia-Munoz, A., Shepardson,
Manson, J. C., et al. (2007). Cellular prion protein regulates beta- N. E., Smith, I., et al. (2008). Amyloid-beta protein dimers
secretase cleavage of the Alzheimer’s amyloid precursor protein. isolated directly from Alzheimer’s brains impair synaptic
Proceedings of the National Academy of Sciences of the United plasticity and memory. Nature Medicine, 14, 837–842.
States of America, 104, 11062–11067. Shaw, L. M., Vanderstichele, H., Knapik-Czajka, M., Clark, C. M.,
Postina, R., Schroeder, A., Dewachter, I., Bohl, J., Schmitt, U., Kojro, Aisen, P. S., Petersen, R. C., et al. (2009). Cerebrospinal fluid
E., et al. (2004). A disintegrin-metalloproteinase prevents biomarker signature in Alzheimer’s disease neuroimaging
amyloid plaque formation and hippocampal defects in an initiative subjects. Annals of Neurology, 65, 403–413.
12 Neuromol Med (2010) 12:1–12

Shimizu, T., Fukuda, H., Murayama, S., Izumiyama, N., & Shirasawa, amyloid precursor protein by the transmembrane aspartic
T. (2002). Isoaspartate formation at position 23 of amyloid beta protease BACE. Science, 286, 735–741.
peptide enhanced fibril formation and deposited onto senile Vetrivel, K. S., Meckler, X., Chen, Y., Nguyen, P. D., Seidah, N. G.,
plaques and vascular amyloids in Alzheimer’s disease. Journal Vassar, R., et al. (2009). Alzheimer disease Abeta production in
of Neuroscience Research, 70, 451–461. the absence of S-palmitoylation-dependent targeting of BACE1
Siemes, C., Quast, T., Kummer, C., Wehner, S., Kirfel, G., Müller, U., to lipid rafts. The Journal of Biological Chemistry, 284, 3793–
et al. (2006). Keratinocytes from APP/APLP2-deficient mice are 3803.
impaired in proliferation, adhesion and migration in vitro. Vogt, D. L., Thomas, D., Galvan, V., Bredesen, D. E., Lamb, B. T. &
Experimental Cell Research, 312, 1939–1949. Pimplikar, S. W. (2009). Abnormal neuronal networks and
Skovronsky, D. M., Moore, D. B., Milla, M. E., Doms, R. W., & Lee, seizure susceptibility in mice overexpressing the APP intracel-
V. M. (2000). Protein kinase C-dependent alpha-secretase lular domain. Neurobiology of Aging (in press).
competes with beta-secretase for cleavage of amyloid-beta Wakabayashi, T., & De Strooper, B. (2008). Presenilins: Members of
precursor protein in the trans-golgi network. The Journal of the gamma-secretase quartets, but part-time soloists too. Phys-
Biological Chemistry, 275, 2568–2575. iology, 23, 194–204.
Sun, B., Zhou, Y., Halabisky, B., Lo, I., Cho, S. H., Mueller-Steiner, Waldron, E., Isbert, S., Kern, A., Jaeger, S., Martin, A. M., Hébert, S.
S., et al. (2008). Cystatin C-cathepsin B axis regulates amyloid S., et al. (2008). Increased AICD generation does not result in
beta levels and associated neuronal deficits in an animal model increased nuclear translocation or activation of target gene
of Alzheimer’s disease. Neuron, 60, 247–257. transcription. Experimental Cell Research, 314, 2419–2433.
Tabuchi, K., Chen, G., Südhof, T. C., & Shen, J. (2009). Conditional Wegiel, J., Kuchna, I., Nowicki, K., Frackowiak, J., Mazur-Kolecka,
forebrain inactivation of nicastrin causes progressive memory B., Imaki, H., et al. (2007). Intraneuronal Abeta immunoreac-
impairment and age-related neurodegeneration. Journal of tivity is not a predictor of brain amyloidosis-beta or neurofibril-
Neuroscience, 29, 7290–7301. lary degeneration. Acta Neuropathologica, 113, 389–402.
Tamgüney, G., Giles, K., Glidden, D. V., Lessard, P., Wille, H., Westmeyer, G. G., Willem, M., Lichtenthaler, S. F., Lurman, G.,
Tremblay, P., et al. (2008). Genes contributing to prion Multhaup, G., Assfalg-Machleidt, I., et al. (2004). Dimerization
pathogenesis. Journal of General Virology, 89, 1777–1788. of beta-site beta-amyloid precursor protein-cleaving enzyme.
Tanabe, C., Hotoda, N., Sasagawa, N., Sehara-Fujisawa, A., Maruy- The Journal of Biological Chemistry, 279, 53205–53212.
ama, K., & Ishiura, S. (2006). ADAM19 is tightly associated Willem, M., Garratt, A. N., Novak, B., Citron, M., Kaufmann, S.,
with constitutive Alzheimer’s disease APP alpha-secretase in Rittger, A., et al. (2006). Control of peripheral nerve myelination
A172 cells. Biochemical and Biophysical Research Communi- by the beta-secretase BACE1. Science, 314, 664–666.
cations, 352, 111–117. Winkler, E., Hobson, S., Fukumori, A., Dümpelfeld, B., Luebbers, T.,
Taylor, C. J., Ireland, D. R., Ballagh, I., Bourne, K., Marechal, N. M., Baumann, K., et al. (2009). Purification, pharmacological
Turner, P. R., et al. (2008). Endogenous secreted amyloid modulation, and biochemical characterization of interactors of
precursor protein-alpha regulates hippocampal NMDA receptor endogenous human gamma-secretase. Biochemistry, 48, 1183–
function, long-term potentiation and spatial memory. Neurobi- 1197.
ology of Diseases, 31, 250–260. Wirths, O., Multhaup, G., Czech, C., Blanchard, V., Moussaoui, S.,
Thathiah, A., Spittaels, K., Hoffmann, M., Staes, M., Cohen, A., Tremp, G., et al. (2001). Intraneuronal Abeta accumulation
Horré, K., et al. (2009). The orphan G protein-coupled receptor 3 precedes plaque formation in beta-amyloid precursor protein and
modulates amyloid-beta peptide generation in neurons. Science, presenilin-1 double-transgenic mice. Neuroscience Letters, 306,
323, 946–951. 116–120.
Thinakaran, G., Borchelt, D. R., Lee, M. K., Slunt, H. H., Spitzer, L., Wolfe, M. S. (2008). Inhibition and modulation fo c-secretase for
Kim, G., et al. (1996). Endoproteolysis of presenilin 1 and Alzheimer’s disease. Neurotherapeutics, 5, 391–398.
accumulation of processed derivatives in vivo. Neuron, 17, 181– Yang, T., Arslanova, D., Gu, Y., Augelli-Szafran, C., & Xia, W.
190. (2008). Quantification of gamma-secretase modulation differen-
Thinakaran, G., & Koo, E. H. (2008). Amyloid precursor protein tiates inhibitor compound selectivity between two substrates
trafficking, processing, and function. The Journal of Biological Notch and amyloid precursor protein. Molecular Brain, 1, 1–15.
Chemistry, 283, 29615–29619. Yang, L. B., Lindholm, K., Yan, R., Citron, M., Xia, W., Yang, X. L.,
Tu, H., Nelson, O., Bezprozvanny, A., Wang, Z., Lee, S. F., Hao, Y. et al. (2003). Elevated beta-secretase expression and enzymatic
H., et al. (2006). Presenilins form ER Ca2? leak channels, a activity detected in sporadic Alzheimer disease. Nature Medi-
function disrupted by familial Alzheimer’s disease-linked muta- cine, 9, 3–4.
tions. Cell, 126, 981–993. Yu, H., Saura, C. A., Choi, S. Y., Sun, L. D., Yang, X., Handler, M.,
Vassar, R., Bennett, B. D., Babu-Khan, S., Kahn, S., Mendiaz, E. A., et al. (2001). APP processing and synaptic plasticity in
Denis, P., et al. (1999). b-Secretase cleavage of Alzheimer’s presenilin-1 conditional knockout mice. Neuron, 31, 713–726.

You might also like