You are on page 1of 13

Journal of Alzheimer’s Disease 19 (2010) 311–323 311

DOI 10.3233/JAD-2009-1221
IOS Press

Mini-Forum Article

Alzheimer’s Disease and the Amyloid-β


Peptide
M. Paul Murphy∗ and Harry LeVine, III∗
Department of Molecular and Cellular Biochemistry and the Sanders-Brown Center on Aging, University of
Kentucky, Lexington, KY, USA

Accepted 3 May 2009

Abstract. Alzheimer’s disease (AD) pathogenesis is widely believed to be driven by the production and deposition of the amyloid-
β peptide (Aβ). For many years, investigators have been puzzled by the weak to nonexistent correlation between the amount
of neuritic plaque pathology in the human brain and the degree of clinical dementia. Recent advances in our understanding of
the development of amyloid pathology have helped solve this mystery. Substantial evidence now indicates that the solubility of
Aβ, and the quantity of Aβ in different pools, may be more closely related to disease state. The composition of these pools of
Aβ reflects different populations of amyloid deposits and has definite correlates with the clinical status of the patient. Imaging
technologies, including new amyloid imaging agents based on the chemical structure of histologic dyes, are now making it
possible to track amyloid pathology along with disease progression in the living patient. Interestingly, these approaches indicate
that the Aβ deposited in AD is different from that found in animal models. In general, deposited Aβ is more easily cleared from
the brain in animal models and does not show the same physical and biochemical characteristics as the amyloid found in AD.
This raises important issues regarding the development and testing of future therapeutic agents.

Keywords: Amyloid-β, amyloid-β protein precursor, fibril, oligomer

INTRODUCTION AβPP) or in presenilin-1 (PS1) or presenilin-2 (PS2).


Either PS1 or PS2 can be the catalytic subunit of γ-
The two hallmark pathologies required for a diag- secretase, which is the final endoprotease in the path-
nosis of Alzheimer’s disease (AD) are the extracellu- way that generates the peptide. Despite this genetic
lar plaque deposits of the amyloid-β peptide (Aβ) and evidence and the demonstrated involvement of Aβ in
the flame-shaped neurofibrillary tangles of the micro- inducing synaptic dysfunction, disrupting neural con-
tubule binding protein tau. Familial early onset forms nectivity, and association with neuronal death in a brain
of AD are associated with mutations either in the pre- region-specific manner, the amounts and distribution
cursor protein for Aβ (the amyloid-β protein precursor, of Aβ deposition are only weakly correlated with the
clinical expression of the disease.
AD is characterized clinically by a progressive and
∗ Address for correspondences: M. Paul Murphy, Department of
gradual decline in cognitive function and neuropatho-
Molecular and Cellular Biochemistry and the Sanders-Brown Center
on Aging University of Kentucky, 800 S. Limestone, Lexington, KY logically by the presence of neuropil threads, specific
40536-0230, USA. Tel.: +1 859 257 1412 x 490; Fax: +1 859 neuron loss, and synapse loss in addition to the hallmark
257 9479; E-mail: mpmurp3@email.uky.edu. Harry LeVine, III, findings of neurofibrillary tangles and senile plaques.
Department of Molecular and Cellular Biochemistry and the Sanders-
The methods used to assess the pathology and classi-
Brown Center on Aging University of Kentucky, 800 S. Limestone,
Lexington, KY 40536-0230, USA. Tel.: +1 859 257 1412 x 224; fy stages of AD have been standardized and codified
Fax: +1 859 323 2866; E-mail: hlevine@email.uky.edu. to provide a guide for clinicians. Standard measures

ISSN 1387-2877/10/$27.50  2010 – IOS Press and the authors. All rights reserved
312 M.P. Murphy and H. LeVine, III / AD Progression and the State of Aβ

of pathology refer to the density of neuritic amyloid results is that Aβ acts as a trigger for a degenerative
plaques and neurofibrillary tangles of tau protein in af- process that continues even if it is removed [3]. It is
fected brain regions. The presence of neuritic plaques not clear what the mechanism might be for this contin-
composed (in large part) of highly insoluble Aβ in the ued degeneration, although a continued accumulation
brain parenchyma is required for a diagnosis of AD. of misfolded hyperphosphorylated tau, leading directly
Deposits of tau protein are also present, although they to further neuron loss, is perhaps the most likely can-
are also found in a number of less common neurode- didate. However, this is a difficult hypothesis to test
generative diseases, notably in the absence of neurit- because it requires the reliable identification of subjects
ic plaques. The neurofibrillary tangles in the different with AD at a very early, preclinical stage, a feat that is
diseases have some distinctive morphological features currently not possible even with the most sensitive and
and may exhibit a distinct composition of tau isoforms dependable means of diagnosing the disease.
that differs from AD [1]. Another possible explanation is that a specific form
Development of a disease stage classification for AD or forms of Aβ are responsible for the massive neuronal
has not been a simple process, nor is there complete death that accompanies the disease. The tools used
consensus with the system(s) that are in place. Defini- to quantify Aβ are not able to distinguish the disease-
tive staging of disease state remains a judgment call de- related Aβ from less relevant forms which weaken the
cided in clinicopathological conferences between clin- correlation with clinical stage. An analogy of this sit-
icians, neuropsychologists, and pathologists. A major uation is found in prion diseases in which the same
deficiency in the staging system is that it can only be protein sequence can assume multiple disease-causing
approximately applied in the living subject. Since AD conformations, each causing neurodegeneration in a
pathology is determined at autopsy, a clinical diagnosis distinct distribution of brain regions resulting in dif-
of probable AD has to be used instead. The lack of an ferent clinical presentations [4,5]. In this review we
in-life diagnostic test greatly hampers research efforts suggest that Aβ is also polymorphic, producing con-
on disease mechanisms and is a particular problem for formational form(s) or specific pool(s) of Aβ that are
clinical trials as it introduces additional heterogeneity disease-relevant while others are less so. Progress is
into the subject population. Therapeutics cannot be being made in methods and systems to delineate these
properly tested if they need to be administered before relevant forms, which will allow testing of this hypoth-
the disease progresses past a certain stage, especially if esis.
this stage is nebulous or the patient population is poorly
defined.
Why Aβ deposition is only weakly related to the de- Aβ METABOLISM, CATABOLISM, AND
gree of dementia has been an enduring puzzle in the CLEARANCE
AD field. While potential floor or ceiling effects in the
amount of Aβ deposition could contribute, there is also The ∼ 4 kDa Aβ peptide, derived from the larger
the possibility that Aβ exerts its major effects early by AβPP, was first isolated as the principal component of
triggering a cascade of processes that, once begun, pro- amyloid deposits in the brain and cerebrovasculature
ceed independently of Aβ. Some support for this argu- of AD and Down’s syndrome patients [6–8]. Although
ment might be found in the human Aβ immunization the function of AβPP itself has not been resolved, ex-
trial (AN-1792). Although the numbers of individuals tensive research has advanced our knowledge of how
to come to autopsy is still very small, the brain Aβ the Aβ peptide is produced, and how it is subsequently
deposition in these cases was far lower than might be degraded within the brain, or transported out into the
expected based on historical levels for a given clinical periphery. The final amount of Aβ that accumulates
stage. In spite of this markedly lower amount of Aβ, as amyloid deposits within the brain is determined by
presumably caused by the immunotherapy, the subjects the interplay of these factors. Changes with disease
continued to decline cognitively to an end stage demen- progression could contribute to the age of disease onset
tia that was clinically indistinguishable from untreated and disease duration.
AD [2]. This is not iron-clad proof that the removal of The enzymatic processes responsible for the metabo-
Aβ succeeded, since we have no way of knowing the lism of AβPP to Aβ are now reasonably well under-
pre-treatment amyloid load, and the number of cases stood. AβPP is sequentially cleaved by two membrane-
is too small for a true cross sectional comparison. It bound endoprotease activities, β- and γ-secretase. β-
is tempting to speculate that the implication of these secretase first cleaves AβPP to release a large secreted
M.P. Murphy and H. LeVine, III / AD Progression and the State of Aβ 313

derivative, sAβPPβ. A fragment of 99 amino acids can be mimicked with a variety of allosteric γ-secretase
(CTFβ, which begins with the N-terminal aspartyl modulating agents [26].
residue of Aβ) remains membrane bound, and is in turn β−Secretase is a membrane-bound aspartyl pro-
rapidly cleaved by γ-secretase to generate Aβ. Cleav- tease, but one that cleaves AβPP and its other sub-
age by γ-secretase is somewhat imprecise, resulting strates outside of the bilayer [27–31]. There are two
in a C-terminal heterogeneity of the resulting peptide major forms of the enzyme, BACE1 and BACE2, which
population. Hence, numerous different Aβ species ex- are > 65% homologous [32,33]. The major form of
ist, but those ending at position 40 (Aβ 40 ) are the most the enzyme responsible for Aβ production, BACE1, is
abundant (∼ 80–90%), followed by 42 (Aβ 42 , ∼5– highly expressed in brain but is also found at lower
10%). The slightly longer forms of Aβ, particularly levels in other organs [27,34]. In contrast, the second
Aβ42 , are more hydrophobic and fibrillogenic, and are form of the enzyme, BACE2, is low in the brain but is
the principal species deposited in the brain [9]. present in most peripheral tissues at higher levels [33].
β−secretase activity is believed to be the rate lim- The knockout of BACE1 in mice leads to a massive
iting step in the amyloidogenic pathway and process- reduction in the levels of the downstream products of
es ∼10% of the total cellular AβPP. The remaining the enzyme (Aβ and CTFβ) in brain [35–37]. Al-
AβPP, close to 90%, is constitutively cleaved by α- though these studies indicate that BACE1 is the ma-
secretase (a collection of metalloprotease enzymes), jor β-secretase activity in brain, some residual activity
generating sAβPPα and the 83 amino acid CTFα. The might be attributable to BACE2 [38], and both forms of
subsequent γ-secretase cleavage of CTFα produces the BACE can compete for substrate [39–43]. β-Secretase
more benign p3 fragment instead of Aβ. γ-Secretase activity and protein are both significantly increased in
cleavage of either membrane bound CTF also gener- sporadic AD [44,46,46]. This effect shows a brain re-
ates a cytosolic element, AICD (AβPP intracellular gional selectivity that roughly parallels disease affect-
domain, sometimes referred to as CTFγ), which may ed regions and is related to both plaque burden and
play a role in signal transduction [10–13]. Because of disease duration [45–47]. β-secretase activity has also
their essential role in the generation of Aβ, both β- and been seen to increase with age in rodents and nonhu-
γ-secretase are considered to be prime targets for the man primates [48], although these species do not devel-
development of anti-AD pharmaceuticals [9,14]. op AD. Recently, evidence has emerged that cathepsin
γ−Secretase is now known to be a multisubunit en- B [49] or cathepsin D [50] may also be able to serve as
zyme composed of the proteins APH1, PEN2, nicas- β-secretase-like enzymes under some circumstances,
trin, and presenilin (PS1 or PS2). The enzyme complex although this view is controversial.
likely contains one copy of each subunit [15] and is Although much emphasis has been placed on un-
responsible for the cleavage of multiple membrane pro- derstanding the production of Aβ from AβPP, in re-
teins in addition to AβPP. Although the exact function- cent years some attention has been shifted to the pro-
al roles of each component have yet to be fully elucidat- cesses responsible for peptide degradation. Two ma-
ed, presenilin is believed to form the active site of the jor enzymes, neprilysin (NEP) and insulin degrading
aspartyl protease [16,17], and nicastrin likely serves as enzyme (also known as insulysin; IDE), are believed
a substrate docking subunit [18]. All four components responsible for most Aβ degradation [51–55]. NEP
are necessary for γ-secretase to mature and function is a plasma membrane bound type II metalloprotease
correctly [19,20]. γ-Secretase has a relatively novel that is responsible for the extracellular degradation of
mechanism in that it cleaves within the lipid bilayer a variety of peptides; IDE, also a metalloprotease, is
and can only process substrates that are first cleaved active both intra- and extracellularly [56–58]. IDE has
by another protease to remove a large ectodomain re- approximately a 20-fold higher affinity for insulin com-
gion [21]. The enzyme does not have identified spe- pared to Aβ, but hydrolyzes insulin at a much slow-
cific sequence requirements for substrate recognition, er rate. Thus, insulin acts as an effective inhibitor of
and cleavage within the membrane is instead controlled the IDE-dependent cleavage of Aβ, which may form
by a variety of other factors, such as the length of the the basis for a link between type II diabetes, hyperin-
transmembrane domain [22,23]. Although the amount sulinemia, and AD [58,59]. In the case of AD, both
of γ-secretase activity does not appear to increase in NEP and IDE decrease in normal aging and in disease-
AD, alterations in γ-secretase activity leading to the affected regions [60,61]. Further, NEP has been shown
production of longer forms of Aβ are the major genetic to decrease in the cerebrospinal fluid (CSF) in early
cause of early onset, familial AD [24,25], an effect that AD [62]. Although most Aβ degradation can be at-
314 M.P. Murphy and H. LeVine, III / AD Progression and the State of Aβ

tributed to NEP and IDE, a substantial body of evidence of the disease. Imaging agents based on analogs of his-
indicates a likely role for lysosomal degradation, by tological dyes which are more discriminating among
enzymes such as cathepsin B [63]. conformational states could then be used antemortem
In spite of substantial catabolism within the brain, to evaluate the pathological components of the disease
a significant amount of Aβ remains undegraded. As stage.
with other metabolites, mechanisms exist to transport Microscopists use the staining characteristics and
Aβ across the blood brain barrier (BBB) and out in- morphology of the lesions in AD to define different
to the circulation. Interfering with this mechanism forms of the deposits. On the other hand, biochemists
causes a large increase in the amount of Aβ that re- look for other ways to differentiate these forms and to
mains in the brain, leading to its ultimate accumula- determine their composition. The silver-stained struc-
tion [64]. Soluble Aβ is exchanged across the BBB by tures in AD brain that also stain with the classic histo-
two principle mechanisms, the low-density lipoprotein logic dyes Congo Red, and Thioflavins S and T are fib-
receptor-related protein (LRP) on the abluminal (brain) rillar proteinaceous structures that are highly insoluble
side [65], and the receptor for advanced glycation end under most conditions. Although these structures are
products (RAGE) on the luminal (blood) side [66]. The composed of a major key protein (Aβ or tau), addition-
net efflux of Aβ across the BBB can predict the degree al proteins and certain glycolipids are also associated
of cerebral amyloid burden [67]. It is unclear why a with them while not being part of the fibril structures
bidirectional mechanism exists for the transport of Aβ themselves. Since these components are also found in
or if this transport has an important physiological role normal brain, they are generally not considered to be
that is unrelated to AD. However, it is possible that pathologic (however, their presence can occasionally
the disruption of these mechanisms, coupled with other interfere with various methods of Aβ detection, par-
extensive co-morbid vascular abnormalities within the ticularly those that are antibody-dependent). Neurit-
AD brain, contribute significantly to and are affected ic plaques (Aβ), cerebrovascular amyloid (Aβ), and
by the development of amyloid pathology [68]. neurofibrillary tangles (tau) can be purified away from
other insoluble components and separated from each
other [69]. They resist solubilization even with harsh
THE CONCEPT OF Aβ POOLS detergents such as sodium dodecyl sulfate (SDS) and
Sarkosyl, requiring concentrated formic acid for de-
A look through a microscope at a silver-stained Sec- polymerization. When the denaturants are removed,
tion of AD brain tissue immediately shows the hetero- the fibrils spontaneously reassemble into their previous
geneity of the pathology. The use of silver impregna- form.
tion techniques to evaluate brain pathology is an old While original efforts were designed to identify the
approach, and the ones in use today differ little from components of the prominent plaques and tangles in
those used by Alois Alzheimer when he initially de- the brain, a series of more finely graded extraction pro-
scribed the disease. Over more than a century, a clas- cedures have been applied to investigate the transition
sification system has evolved to make sense of the pro- of the Aβ peptide from soluble monomer through dif-
gression of the disease, a tour-de-force effort of count- fuse plaques and deposits into neuritic plaques. Low
less clinicians and pathologists. However, in the pro- ionic strength alkaline solutions (such as low salt di-
cess a certain amount of information was averaged out ethylamine) and SDS-treatment can extract fractions of
and lost. In the past, simplification was necessary to Aβ of intermediate solubility, and 70% formic acid dis-
improve understanding and provide a basis for test- solves the remainder (mostly neuritic plaques), leaving
ing hypotheses of the disease mechanism. Modern ad- lipofuscin granules but no Aβ in the small amount of
vances in technology, as well as new ways of studying residue [70]. Tau protein in the AD brain is character-
the relationship of the pathology to brain biology and ized as pools of Sarkosyl-soluble and -insoluble mate-
our improved understanding of the biochemical basis rial, with the latter thought to represent the neurofibril-
of the disease, require a re-evaluation of the ‘lumping’ lary tangles which require strong acid for dissociation
procedure used to discriminate among subpopulations into monomeric tau [71].
of disease cases and different subtypes of plaques and From studies of the extractability of Aβ at differ-
tangles. If these misfolded protein structures are in- ent stages of AD progression compared to the histol-
deed polymorphic like prions, specific subpopulations ogy, aqueous buffer or dilute alkali released soluble
will be strongly correlated with the clinical progression and adsorbed Aβ species, SDS-extraction removed dif-
M.P. Murphy and H. LeVine, III / AD Progression and the State of Aβ 315

fuse deposits, and formic acid extraction was required pick up early stage disease or predict progression to
to solubilize the neuritic plaques and cerebrovascular AD before clinical symptoms appear. This has proven
amyloid [72,73]. Brain tissue from AβPP/PS1 knock- very difficult to sort out and remains an area of in-
in mice at corresponding stages of Aβ deposition pro- tense investigation [79]. An alternative approach using
duced analogous extraction profiles. Hence, at this analogs of amyloid dyes to image and quantify amy-
level of resolution, more distinct than staining mor- loid pathology in the brain of subjects has produced
phology, the Aβ peptide forms structures with similar exciting results and in the process perhaps has provid-
physical-chemical properties in AD and in mouse mod- ed insight into differences between the human disease
els of brain Aβ pathology. Tau-containing structures and the systems used to recapitulate the process in an-
remain to be characterized in this way. imal models. While oligomeric Aβ is likely present at
Immunohistochemical staining of tissue Sections re- concentrations far below the detection limits of current
veals numerous deposits of Aβ peptide with few or imaging technology, other pools of Aβ may be more
no fibrils that stain poorly or not at all with amyloid suitable for imaging. The SDS-insoluble Aβ isolat-
dyes [74]. Although this diffuse amyloid was once ed from AD brain has shown to have a distinct fibril
thought to be an early stage of neuritic plaque devel- molecular structure by solid state 13 C NMR and could
opment, this has proven difficult to resolve conclusive- be a target for development of an imaging ligand [80].
ly. The amyloid dyes (Congo Red and the Thioflavins) A derivative of the amyloid dye, Thioflavin T, Pitts-
light up neuritic plaques but only weakly react with burgh Compound 1 (PIB, 2-(4’-methylaminophenyl)-
other deposits that can be silver stained in brain Sec- 6-hydroxybenzothiazole) was prepared with physico-
tions. Congo red birefringence reveals oriented period- chemical properties that made it a good brain imaging
ic organization in neuritic plaques but not in these oth- ligand [81]. After labeling with 11 C for PET imag-
er structures, and this diffuse material has been shown ing, increasing amounts of deposition in specific brain
to be Aβ peptide by sequence-specific immunostain- regions could be detected in parallel with decreased
ing. The lack of birefringence is due to a lack of reg- glucose metabolism ( 18 F-fluorodeoxyglucose) in those
ular fibril structure. Conformation-dependent antibod- same regions following the disease progression in AD
ies that do not recognize diffuse amyloid deposits, but patients [82]. The utility of this and other probes for
which recognize synthetic fibrils and neuritic plaques detecting MCI and predicting which of those subjects
have been described [75,76]. Not all forms of Aβ fib- will progress to AD is being evaluated [82,83]. How-
rils react equally well with all anti-fibril monoclonal ever, PIB has already, from the point of view of this re-
antibodies, suggestive of the ability of such reagents to view, provided a valuable perspective on potential Aβ
discriminate polymorphic fibril forms, but this property polymorphism and a possible explanation for why our
remains to be conclusively demonstrated. animal models are Aβ pathology models but fail to re-
Detailed study of neuritic plaques stained with a capitulate the full spectrum of AD pathology with its
series of fluorescent polythiophene derivatives whose massive cell death.
emission spectra are sensitive to the amount of order Histological staining with dyes takes place at high
in an amyloid fibril revealed that individual plaques or concentrations and then the tissue is washed (differenti-
regions within a single plaque could be differently or- ated) to remove excess dye. In brain imaging, for tech-
ganized [77]. Synthetic Aβ peptide fibrils prepared un- nical and toxicology reasons, only nanomolar concen-
der different conditions (agitation or quiescence) stain trations of ligand can be used. PIB is employed similar
differentially with these probes indicating that Aβ fib- to a pharmacological ligand in these studies. There are
ril polymorphism can readily occur. Synthetic fibril both high (nM) and low (µM) affinity PIB binding sites
structural polymorphism under different fibril-forming on synthetic and biological Aβ fibrils [84]. Only the
conditions is also observed by solid state NMR [78]. high affinity binding site is significantly occupied un-
Assessing the development of AD pathology in the der imaging conditions. When binding studies of PIB
living human brain has been a dream of clinicians. A to AD brain fibrils are performed a large proportion of
means to study AD in vivo serves minimally as a sur- the binding is due to the high affinity site. By contrast,
rogate measure of disease progression, and hopefully synthetic Aβ fibrils, aged transgenic mouse Aβ brain as
as a diagnostic tool to detect an early, preclinical stage well as the brains of aged non-human primates (squir-
of the disease. A great deal of effort has gone into rel monkey, macaques, chimpanzee) [85], all with hu-
the development of noninvasive, sensitive and specif- man sequence Aβ peptide in similar amounts, have the
ic biomarkers in the blood or CSF for AD that could low affinity site in overwhelming proportion. Images
316 M.P. Murphy and H. LeVine, III / AD Progression and the State of Aβ

can only be obtained in transgenic mice when PIB with systems. The explanation for this difference in stabil-
ten-fold higher specific radioactivity is used to detect ity is unknown. Aβ oligomers with SDS-stable sub-
the very small amount of high affinity binding site [86]. structures as small as dimers isolated from AD brain
Although other explanations are currently being tested, and CSF have been shown to disrupt synaptic electro-
one possibility is that the polymorphic form of Aβ fibril physiology [95,96]. Whether these intermediates arise
or Aβ-containing complex in AD is enriched in high during assembly, or after disassembly in vivo, remains
affinity PIB sites. to be determined. Soluble synthetic Aβ oligomers are
Another indication of the unique polymorphic struc- highly polymorphic with stable sizes that depend on the
ture of AD brain Aβ is that it is much more effi- method of preparation [97]. While investigators agree
cient at seeding Aβ fibril formation when injected in- that soluble oligomers are biologically active and can
to the brains of transgenic mice producing Aβ peptide cause cell death under some conditions, the mode of
than are equivalent amounts of synthetic Aβ fibrils or action of soluble oligomers also remains to be settled.
Aβ extracts from plaque-containing transgenic mouse Receptor-mediated effects are noted [98,99] as well as
brains [87–89]. direct activity of oligomers on the membrane bilayer,
especially at high (µM) concentrations which may stem
from their surface activity. The concentration of solu-
ble oligomers in CSF or brain interstitial fluid is in the
MULTIMERIZATION, NUCLEATION, AND
pM range [100].
DEPOSITION
Fibril formation is studied at high micromolar con-
centrations of monomeric synthetic peptide to increase
The assembly of Aβ into multimeric structures is the probability of a fibril nucleus forming. There is ev-
key to the biological effect of those species. There are idence [101] that soluble oligomer formation and fibril
two phases of assembly which have different charac- formation may be different pathways, although mech-
teristics and lead to assemblies with different biolog- anistically both processes have to pass through multi-
ical properties. Initial work with Aβ focused on the meric stages. Since Aβ concentrations in brain inter-
histological hallmarks of AD, the amorphous and fib- stitial fluid are at least three-orders of magnitude low-
rillar deposits of the peptide. Current focus is on the er than in fibril forming assays, it is likely that fibril
earlier phase of Aβ assembly which involves soluble formation is nucleated on extracellular matrix or cell
multimers of the peptide. These structures are orders of surfaces. Fibril growth by extension on both synthetic
magnitude more toxic to cells of different types than are and AD brain pre-existing fibrils is linearly dependent
the fibrils and trigger a different set of toxic events [90]. on the Aβ monomer concentration [102] and is high-
They are also morphologically and conformationally ly specific for the form of amyloid fibril [103]. The
distinct. Oligomer-specific conformational antibodies process is reversible in vivo in transgenic mouse mod-
do not recognize monomer or fibrils, and fibril-specific els monitored by multiphoton microscopy [104] and
antibodies do not recognize soluble oligomers. While is surprisingly rapid in that system and includes vas-
total abandonment of fibril involvement in AD at this cular amyloid [105]. Although this process has not
time is probably premature given the PIB story dis- been documented in the live human brain, the effects
cussed earlier and the potential for plaque involvement of active and passive antibody administration in animal
with oligomer populations [91], the focus of the Aβ models [106] and human immunization trials [2] sug-
field has shifted to soluble oligomers. gest that Aβ deposits in the brain will be in equilibrium
Oligomers form readily from the Aβ 1−42 peptide, with the interstitial Aβ. There is also evidence consis-
less well so from the more abundant Aβ 1−40 [92]. tent with insoluble Aβ deposits serving as a reservoir
There is a close correlation between the ratio of 42/40 for soluble oligomers [91]. Not all pools of Aβ in the
and age of disease onset in familial AD [93]. The human brain may participate in this rapid exchange of
C-terminus of Aβ1−42 is critical for oligomer forma- monomer and the relative involvement of the different
tion. Bitan and colleagues [94] defined some of the pools in the pathologic process is unknown.
structural parameters for the different steps of in vit-
ro oligomer assembly with synthetic peptides mutated MODELING STATES OF Aβ AND
in that region. While the early intermediates during ALZHEIMER’S DISEASE
oligomerization of synthetic peptide are unstable and
require photochemical trapping of the intermediates, AD is a human specific disorder. Even our most
stable small oligomers can be isolated from biological closely related primate relatives do not develop pathol-
M.P. Murphy and H. LeVine, III / AD Progression and the State of Aβ 317

ogy, much less the clinical outcome that can be consid- in human disease. Thus, canines may represent a use-
ered to be actual AD. Still, much of our understanding ful intermediate between genetically modified mouse
of how amyloid pathology develops has been driven by models and AD. In this regard it is worth noting that
studies in a variety of animal models. Although there immunization with fibrillar Aβ in aged canines may be
is no animal model that accurately reflects every facet a better parallel to human trials with this therapeutic
of AD, there are many models of Aβ deposition. In approach than preclinical mouse models, although data
a very broad sense, these can be subdivided into mod- from humans are still sparse [2,114].
els where amyloid pathology develops naturally with Insights from mouse models have been indispensible
age, and genetically modified mice that express mutant for our understanding of amyloid deposition in vivo.
forms of AβPP. Animals in which amyloid deposition It is beyond the scope of this article to review the ex-
occurs naturally is attractive in that the researcher is not tensive literature on mouse models of Aβ deposition
burdened with the numerous caveats that accompany and AD pathology, and several excellent reviews ex-
genetically modified mice, such as separating the con- ist [115–117]. There are a number of points worth men-
tributions of overexpression and introduced mutations tioning. First, although mouse models have economics
to the model phenotype. However, animals in which and speed as a major advantage, with some models de-
amyloid deposition occurs as a consequence of normal veloping amyloid deposition at birth [118], all models
aging have a substantial longer lifespan than rodents require the introduction of some combination of famil-
(very long in the case of some nonhuman primates), ial AD mutations into AβPP or PS1 or both. As a gen-
and their use can be difficult to justify based purely on eral rule, incorporating more mutations accelerates the
their associated cost. pathology. The hidden cost in this endeavor is that sub-
Although nonhuman primates (NHPs) have identical stantial alterations are being introduced into proteins
Aβ sequence to humans and a near identical AβPP se- (particularly AβPP) with unknown function, and the
quence, and they overlap in many aspects with relevant consequences of these mutations beyond driving amy-
human biochemical pathways, they develop surpris- loid deposition are unknown. Second, with few ex-
ingly little AD-like neuropathology with age. While ceptions [73,119], it is necessary to overexpress AβPP
older NHPs typically show small amounts of amyloid containing human sequence Aβ at relatively high lev-
deposition, this is quite modest compared to cases of els (using an ectopic promoter) to drive the deposition
AD [107–109]. Although abnormal neurofilaments can of Aβ. This carries with it the same note of caution
frequently be identified with some amyloid deposits, as does the introduction of mutations. An additional
neurofibrillary tangle pathology is not a typical feature concern is that in the majority of these models the ro-
of pathology in NHPs [108], although chimpanzees dent Aβ is present which can affect the assembly of the
may be an exception [110]. Only a small amount of human sequence peptide in vitro and in vivo [120–122].
biochemical work has been performed on amyloid iso- Caveats aside, insights derived from genetically
lated from NHPs, although interestingly, a recent study modified mice have been highly useful. Work in trans-
indicated that there may be more soluble Aβ peptide in genic mice showed that amyloid deposition is driven
the chimpanzee than in AD [110]. almost entirely by Aβ 42 , and not Aβ40 [123]. Trans-
Aged canines also develop substantial amyloid depo- genic mice have been used to demonstrate convincing-
sition with age. Unlike aged NHPs, which may require ly that Aβ increases the rate of neurofibrillary tangle
several decades, canines show substantial amyloid de- pathology in mice which also express mutant tau pro-
position from approximately ten years of age [111]. tein [124,125], placing Aβ pathology firmly upstream
Amyloid deposition in canines is also correlated with of tangle pathology in the hierarchy of disease progres-
age-related cognitive dysfunction [112], although little sion. Higher order, soluble oligomeric forms of Aβ are
neuronal loss is observed. Similar to AD, Aβ deposi- toxic to neurons and cause deficits in long term potenti-
tion in canines is driven by Aβ 42 ; however, the deposi- ation [126,127], providing crucial evidence that a solu-
tion occurs almost exclusively in diffuse deposits, with ble intermediate form of Aβ may drive the early disease
no neuritic plaques or neurofibrillary tangle patholo- process rather than the amyloid deposits themselves.
gy [113]. Nonetheless, compared to NHPs, the quan- Recently, mice have also been used to demonstrate that
tity of deposited Aβ is comparable to AD cases, and a amyloid can deposit with extraordinary rapidity in the
substantial proportion is highly insoluble [114]. This brain [104]. What is also remarkable is the large num-
would seem to suggest that the amyloid in the aged ca- ber of preclinical interventions that have been shown
nine quantitatively overlaps with the amount of amyloid to reverse amyloid deposition in mice. This may be a
318 M.P. Murphy and H. LeVine, III / AD Progression and the State of Aβ

function of the state of the amyloid in mice which is posed for clinical imaging studies may not be report-
consistently less crosslinked and chemically modified ing the same form of Aβ, much less a disease-related
than in AD [128]. The amyloid deposited in the mouse one. The uncritical use of such agents could potentially
brain may be considerably more plastic than human generate further confusion in the literature. On the oth-
amyloid, possibly as a consequence of a far shorter in er hand, these ligands could be quite useful, provided
vivo dwell time in mice, thus avoiding the extensive that their binding site selectivity has been characterized
Aβ modification and cross-linking observed in human under in vivo imaging-like conditions.
material.
The observation that the stoichiometry of high affin-
ity PIB-binding in AD brain is drastically reduced in ACKNOWLEDGMENTS
the transgenic AβPP mouse model [84] may be a fur-
ther reflection of the differences between polymorphic Supported by the National Institutes of Health,
forms or complexes of the Aβ amyloid, some of which NS058382 (M.P.M.) and AG005119 (M.P.M. and
may be more related to disease pathology than others. H.L.). The authors would like to thank Dr. Elizabeth
As of yet, it is not known what significance these dif- Head for helpful discussion, and Paula Thomason for
ferences may entail for our understanding of the pro- her expertise in editing the final manuscript.
gression of the disease but they will likely complicate Authors’ disclosures available online (http://www.j-
the development of anti-Aβ targeted therapeutics. alz.com/disclosures/view.php?id=63).

CONCLUSIONS REFERENCES
Understanding of the state of Aβ is of more than [1] Lee VM, Goedert M, Trojanowski JQ (2001) Neurodegener-
academic interest. Clinical concepts have not kept pace ative tauopathies. Annu Rev Neurosci 24, 1121-1159.
with molecular knowledge and remain heavily depen- [2] Holmes C, Boche D, Wilkinson D, Yadegarfar G, Hopkins V,
dent on classical pathology. The difficulty in defin- Bayer A, Jones RW, Bullock R, Love S, Neal JW, Zotova E,
Nicoll JA (2008) Long-term effects of Abeta42 immunisation
ing clinically useful biomarkers of AD progression in- in Alzheimer’s disease: follow-up of a randomised, placebo-
dicates that new parameters need to be investigated, controlled phase I trial. Lancet 372, 216-223.
which could include particular molecular polymorphic [3] Holtzman DM (2008) Alzheimer’s disease: Moving towards
a vaccine. Nature 454, 418-420.
forms of Aβ. [4] Collinge J, Clarke AR (2007) A general model of prion strains
One area in which it is important to consider dif- and their pathogenicity. Science 318, 930-936.
ferent states or pools of Aβ is in the development and [5] Aguzzi A, Sigurdson C, Heikenwaelder M (2008) Molecular
use of imaging markers. What has become clear from mechanisms of prion pathogenesis. Annu Rev Pathol 3, 11-
40.
the studies with PIB and similar benzothiazole radioli- [6] Glenner GG, Wong CW (1984) Alzheimer’s disease and
gands is that there are multiple binding sites on Aβ Down’s syndrome: sharing of a unique cerebrovascular amy-
fibrils that have different molecular specificities [129– loid fibril protein. Biochem Biophys Res Commun 122, 1131-
131]. The sites have different stoichiometries on fibrils 1135.
[7] Masters CL, Multhaup G, Simms G, Pottgiesser J, Martins
for their ligands and some appear to partially overlap. RN, Beyreuther K (1985) Neuronal origin of a cerebral amy-
Thus, the issue becomes a pharmacological one. PIB loid: neurofibrillary tangles of Alzheimer’s disease contain
turns out to bind with high affinity to a site distinct from the same protein as the amyloid of plaque cores and blood
binding site(s) for Congo Red derivatives and DDNP vessels. EMBO J 4, 2757-2763.
[8] Masters CL, Simms G, Weinman NA, Multhaup G, McDon-
derivatives. This high affinity PIB site is found at high ald BL, Beyreuther K (1985) Amyloid plaque core protein in
stoichiometry with respect to Aβ in affected regions of Alzheimer disease and Down syndrome. Proc Natl Acad Sci
AD brain. Only very low stoichiometry PIB binding is U S A 82, 4245-4249.
seen in unaffected regions of AD brain or any region of [9] Selkoe DJ (2001) Alzheimer’s disease: genes, proteins, and
therapy. Physiol Rev 81, 741-766.
the plaque-rich transgenic mouse brain (Klunk, Rosen, [10] Cao X, Sudhof TC (2001) A transcriptively active complex of
LeVine, and Walker, unpublished results) or in non- APP with Fe65 and histone acetyltransferase Tip60. Science
human primate temporal or parietal cortex containing 293, 115-120.
[11] Gao Y, Pimplikar SW (2001) The gamma-secretase-cleaved
similar amounts of insoluble Aβ [85]. The type of amy-
C-terminal fragment of amyloid precursor protein mediates
loid binding ligand used is important. The plethora of signaling to the nucleus. Proc Natl Acad Sci U S A 98, 14979-
new imaging molecules with different structures pro- 14984.
M.P. Murphy and H. LeVine, III / AD Progression and the State of Aβ 319

[12] Kimberly WT, Zheng JB, Guenette SY, Selkoe DJ (2001) The [27] Vassar R, Bennett BD, Babu-Khan S, Kahn S, Mendiaz EA,
intracellular domain of the beta-amyloid precursor protein Denis P, Teplow DB, Ross S, Amarante P, Loeloff R, Luo
is stabilized by Fe65 and translocates to the nucleus in a Y, Fisher S, Fuller J, Edenson S, Lile J, Jarosinski MA,
notch-like manner. J Biol Chem 276, 40288-40292. Biere AL, Curran E, Burgess T, Louis JC, Collins F, Tre-
[13] Hardy J, Selkoe DJ (2002) The amyloid hypothesis of anor J, Rogers G, Citron M (1999) Beta-secretase cleavage
Alzheimer’s disease: progress and problems on the road to of Alzheimer’s amyloid precursor protein by the transmem-
therapeutics. Science 297, 353-356. brane aspartic protease BACE. Science 286, 735-741.
[14] Rochette MJ, Murphy MP (2002) Gamma-Secretase: sub- [28] Hussain I, Powell D, Howlett DR, Tew DG, Meek TD, Chap-
strates and inhibitors. Mol Neurobiol 26, 81-95. man C, Gloger IS, Murphy KE, Southan CD, Ryan DM,
[15] Sato T, Diehl TS, Narayanan S, Funamoto S, Ihara Y, De Smith TS, Simmons DL, Walsh FS, Dingwall C, Christie G
Strooper B, Steiner H, Haass C, Wolfe MS (2007) Active (1999) Identification of a Novel Aspartic Protease (Asp 2) as
gamma-secretase complexes contain only one of each com- beta-Secretase. Mol Cell Neurosci 14, 419-427.
ponent. J Biol Chem 282, 33985-33993. [29] Sinha S, Anderson JP, Barbour R, Basi GS, Caccavello R,
[16] Wolfe MS, Xia W, Ostaszewski BL, Diehl TS, Kimber- Davis D, Doan M, Dovey HF, Frigon N, Hong J, Jacobson-
ly WT, Selkoe DJ (1999) Two transmembrane aspartates Croak K, Jewett N, Keim P, Knops J, Lieberburg I, Power M,
in presenilin-1 required for presenilin endoproteolysis and Tan H, Tatsuno G, Tung J, Schenk D, Seubert P, Suomensaari
gamma-secretase activity. Nature 398, 513-517. SM, Wang S, Walker D, John V, et al. (1999) Purification
[17] Li YM, Xu M, Lai MT, Huang Q, Castro JL, DiMuzio-Mower and cloning of amyloid precursor protein beta-secretase from
J, Harrison T, Lellis C, Nadin A, Neduvelil JG, Register human brain. Nature 402, 537-540.
RB, Sardana MK, Shearman MS, Smith AL, Shi XP, Yin [30] Yan R, Bienkowski MJ, Shuck ME, Miao H, Tory MC, Pauley
KC, Shafer JA, Gardell SJ (2000) Photoactivated gamma- AM, Brashier JR, Stratman NC, Mathews WR, Buhl AE,
secretase inhibitors directed to the active site covalently label Carter DB, Tomasselli AG, Parodi LA, Heinrikson RL, Gur-
presenilin 1. Nature 405, 689-694. ney ME (1999) Membrane-anchored aspartyl protease with
[18] Shah S, Lee SF, Tabuchi K, Hao YH, Yu C, LaPlant Q, Ball Alzheimer’s disease beta-secretase activity. Nature 402, 533-
H, Dann CE, 3rd, Sudhof T, Yu G (2005) Nicastrin functions 537.
as a gamma-secretase-substrate receptor. Cell 122, 435-447. [31] Lin X, Koelsch G, Wu S, Downs D, Dashti A, Tang J
[19] Kimberly WT, LaVoie MJ, Ostaszewski BL, Ye W, Wolfe (2000) Human aspartic protease memapsin 2 cleaves the beta-
MS, Selkoe DJ (2003) Gamma-secretase is a membrane pro- secretase site of beta-amyloid precursor protein. Proc Natl
tein complex comprised of presenilin, nicastrin, Aph-1, and Acad Sci U S A 97, 1456-1460.
Pen-2. Proc Natl Acad Sci U S A 100, 6382-6387. [32] Saunders AJ, Kim T-W, Tanzi RE, Fan W, Bennett BD, Sa-
[20] Edbauer D, Winkler E, Regula JT, Pesold B, Steiner H, Haass fura B-K, Luo Y, Louis J-C, McCaleb M, Citron M, Vas-
C (2003) Reconstitution of gamma-secretase activity. Nat sar R, Richards WG (1999) BACE maps to chromosome 11
Cell Biol 5, 486-488. and a BACE homolog, BACE2, reside in the obligate Down
[21] Selkoe DJ, Wolfe MS (2007) Presenilin: running with scis- syndrome region of chromosome 21. Science 286, 1255.
sors in the membrane. Cell 131, 215-221. [33] Bennett BD, Babu-Khan S, Loeloff R, Louis JC, Curran E,
[22] Murphy MP, Hickman LJ, Eckman CB, Uljon SN, Wang R, Citron M, Vassar R (2000) Expression analysis of BACE2 in
Golde TE (1999) gamma-Secretase, evidence for multiple brain and peripheral tissues. J Biol Chem 275, 20647-20651.
proteolytic activities and influence of membrane positioning [34] Marcinkiewicz M, Seidah NG (2000) Coordinated expres-
of substrate on generation of amyloid beta peptides of varying sion of beta-amyloid precursor protein and the putative beta-
length. J Biol Chem 274, 11914-11923. secretase BACE and alpha-secretase ADAM10 in mouse and
[23] Lichtenthaler SF, Beher D, Grimm HS, Wang R, Shearman human brain. J Neurochem 75, 2133-2143.
MS, Masters CL, Beyreuther K (2002) The intramembrane [35] Cai H, Wang Y, McCarthy D, Wen H, Borchelt DR, Price
cleavage site of the amyloid precursor protein depends on the DL, Wong PC (2001) BACE1 is the major beta-secretase for
length of its transmembrane domain. Proc Natl Acad Sci U S generation of Abeta peptides by neurons. Nat Neurosci 4,
A 99, 1365-1370. 233-234.
[24] Scheuner D, Eckman C, Jensen M, Song X, Citron M, Suzuki [36] Luo Y, Bolon B, Kahn S, Bennett BD, Babu-Khan S, Denis
N, Bird TD, Hardy J, Hutton M, Kukull W, Larson E, Levy- P, Fan W, Kha H, Zhang J, Gong Y, Martin L, Louis J-
Lahad E, Viitanen M, Peskind E, Poorkaj P, Schellenberg C, Yan Q, Richards WG, Citron M, Vassar R (2001) Mice
G, Tanzi R, Wasco W, Lannfelt L, Selkoe D, Younkin S deficient in BACE1, the Alzheimer’s beta-secretase, have
(1996) Secreted amyloid beta-protein similar to that in the normal phenotype and abolished beta-amyloid generation.
senile plaques of Alzheimer’s disease is increased in vivo by Nat Neurosci 4, 231-232.
the presenilin 1 and 2 and APP mutations linked to familial [37] McConlogue L, Buttini M, Anderson JP, Brigham EF, Chen
Alzheimer’s disease. Nat Med 2, 864-870. KS, Freedman SB, Games D, Johnson-Wood K, Lee M,
[25] Suzuki N, Cheung TT, Cai X-D, Odaka A, Otvos L, Eckman Zeller M, Liu W, Motter R, Sinha S (2007) Partial reduc-
C, Golde TE, Younkin SG (1994) An increased percentage of tion of BACE1 has dramatic effects on Alzheimer plaque and
long amyloid protein is secreted by familial amyloid protein synaptic pathology in APP Transgenic Mice. J Biol Chem
precursor (APP717 ) mutants. Science 264, 1336-1340. 282, 26326-26334.
[26] Kukar T, Murphy MP, Eriksen JL, Sagi SA, Weggen S, Smith [38] Dominguez D, Tournoy J, Hartmann D, Huth T, Cryns K,
TE, Ladd T, Khan MA, Kache R, Beard J, Dodson M, Merit Deforce S, Serneels L, Camacho IE, Marjaux E, Craessaerts
S, Ozols VV, Anastasiadis PZ, Das P, Fauq A, Koo EH, Golde K, Roebroek AJ, Schwake M, D’Hooge R, Bach P, Kalinke
TE (2005) Diverse compounds mimic Alzheimer disease- U, Moechars D, Alzheimer C, Reiss K, Saftig P, De Strooper
causing mutations by augmenting Abeta42 production. Nat B (2005) Phenotypic and biochemical analyses of BACE1-
Med 11, 545-550. and BACE2-deficient mice. J Biol Chem 280, 30797-30806.
320 M.P. Murphy and H. LeVine, III / AD Progression and the State of Aβ

[39] Hussain I, Powell DJ, Howlett DR, Chapman GA, Gilmour [54] Miller BC, Eckman EA, Sambamurti K, Dobbs N, Chow
L, Murdock PR, Tew DG, Meek TD, Chapman C, Schneider KM, Eckman CB, Hersh LB, Thiele DL (2003) Amyloid-beta
K, Ratcliffe SJ, Tattersall D, Testa TT, Southan C, Ryan peptide levels in brain are inversely correlated with insulysin
DM, Simmons DL, Walsh FS, Dingwall C, Christie G (2000) activity levels in vivo. Proc Natl Acad Sci U S A 100, 6221-
ASP1 (BACE2) cleaves the amyloid precursor protein at the 6226.
beta- secretase site. Mol Cell Neurosci 16, 609-619. [55] Huang SM, Mouri A, Kokubo H, Nakajima R, Suemo-
[40] Farzan M, Schnitzler CE, Vasilieva N, Leung D, Choe H to T, Higuchi M, Staufenbiel M, Noda Y, Yamaguchi H,
(2000) BACE2, a beta-secretase homolog, cleaves at the beta- Nabeshima T, Saido TC, Iwata N (2006) Neprilysin-sensitive
site and within the amyloid-beta region of the amyloid-beta synapse-associated amyloid-beta peptide oligomers impair
precursor protein. Proc Natl Acad Sci U S A 97, 9712-9717. neuronal plasticity and cognitive function. J Biol Chem 281,
[41] Yan R, Munzner JB, Shuck ME, Bienkowski MJ (2001) 17941-17951.
BACE2 functions as an alternative alpha-secretase in cells. J [56] Qiu WQ, Walsh DM, Ye Z, Vekrellis K, Zhang J, Podlisny
Biol Chem 276, 34019-34027. MB, Rosner MR, Safavi A, Hersh LB, Selkoe DJ (1998)
[42] Basi G, Frigon N, Barbour R, Doan T, Gordon G, Mc- Insulin-degrading enzyme regulates extracellular levels of
Conlogue L, Sinha S, Zeller M (2003) Antagonistic effects of amyloid beta-protein by degradation. J Biol Chem 273,
beta-site amyloid precursor protein-cleaving enzymes 1 and 32730-32738.
2 on beta-amyloid peptide production in cells. J Biol Chem [57] Vekrellis K, Ye Z, Qiu WQ, Walsh D, Hartley D, Chesneau V,
278, 31512-31520. Rosner MR, Selkoe DJ (2000) Neurons regulate extracellular
[43] Sun X, Wang Y, Qing H, Christensen MA, Liu Y, Zhou W, levels of amyloid beta-protein via proteolysis by insulin-
Tong Y, Xiao C, Huang Y, Zhang S, Liu X, Song W (2005) degrading enzyme. J Neurosci 20, 1657-1665.
Distinct transcriptional regulation and function of the human [58] Qiu WQ, Folstein MF (2006) Insulin, insulin-degrading en-
BACE2 and BACE1 genes. FASEB J 19, 739-749. zyme and amyloid-beta peptide in Alzheimer’s disease: re-
[44] Holsinger RM, McLean CA, Beyreuther K, Masters CL, Evin view and hypothesis. Neurobiol Aging 27, 190-198.
G (2002) Increased expression of the amyloid precursor beta- [59] Luchsinger JA, Tang MX, Shea S, Mayeux R (2004) Hy-
secretase in Alzheimer’s disease. Ann Neurol 51, 783-786. perinsulinemia and risk of Alzheimer disease. Neurology 63,
[45] Fukumoto H, Cheung BS, Hyman BT, Irizarry MC (2002) 1187-1192.
beta-Secretase Protein and Activity Are Increased in the Neo- [60] Caccamo A, Oddo S, Sugarman MC, Akbari Y, LaFerla
cortex in Alzheimer Disease. Arch Neurol 59, 1381-1389. FM (2005) Age- and region-dependent alterations in Abeta-
[46] Yang LB, Lindholm K, Yan R, Citron M, Xia W, Yang XL, degrading enzymes: implications for Abeta-induced disor-
Beach T, Sue L, Wong P, Price D, Li R, Shen Y (2003) ders. Neurobiol Aging 26, 645-654.
Elevated beta-secretase expression and enzymatic activity [61] Miners JS, Van Helmond Z, Chalmers K, Wilcock G, Love
detected in sporadic Alzheimer disease. Nat Med 9, 3-4. S, Kehoe PG (2006) Decreased expression and activity of
[47] Li R, Lindholm K, Yang LB, Yue X, Citron M, Yan R, Beach neprilysin in Alzheimer disease are associated with cerebral
T, Sue L, Sabbagh M, Cai H, Wong P, Price D, Shen Y (2004) amyloid angiopathy. J Neuropathol Exp Neurol 65, 1012-
Amyloid beta peptide load is correlated with increased beta- 1021.
secretase activity in sporadic Alzheimer’s disease patients. [62] Maruyama M, Higuchi M, Takaki Y, Matsuba Y, Tanji
Proc Natl Acad Sci U S A 101, 3632-3637. H, Nemoto M, Tomita N, Matsui T, Iwata N, Mizukami
[48] Fukumoto H, Rosene DL, Moss MB, Raju S, Hyman BT, H, Muramatsu S, Ozawa K, Saido TC, Arai H, Sasaki H
Irizarry MC (2004) Beta-secretase activity increases with (2005) Cerebrospinal fluid neprilysin is reduced in prodromal
aging in human, monkey, and mouse brain. Am J Pathol 164, Alzheimer’s disease. Ann Neurol 57, 832-842.
719-725. [63] Sun B, Zhou Y, Halabisky B, Lo I, Cho SH, Mueller-Steiner
[49] Hook VY, Kindy M, Hook G (2008) Inhibitors of cathepsin S, Devidze N, Wang X, Grubb A, Gan L (2008) Cystatin C-
B improve memory and reduce beta-amyloid in transgenic cathepsin B axis regulates amyloid beta levels and associated
Alzheimer disease mice expressing the wild-type, but not the neuronal deficits in an animal model of Alzheimer’s disease.
Swedish mutant, beta-secretase site of the amyloid precursor Neuron 60, 247-257.
protein. J Biol Chem 283, 7745-7753. [64] Van Uden E, Mallory M, Veinbergs I, Alford M, Rockenstein
[50] Schechter I, Ziv E (2008) Kinetic properties of cathepsin E, Masliah E (2002) Increased extracellular amyloid deposi-
D and BACE 1 indicate the need to search for additional tion and neurodegeneration in human amyloid precursor pro-
beta-secretase candidate(s). Biol Chem 389, 313-320. tein transgenic mice deficient in receptor-associated protein.
[51] Iwata N, Tsubuki S, Takaki Y, Watanabe K, Sekiguchi J Neurosci 22, 9298-9304.
M, Hosoki E, Kawashima-Morishima M, Lee HJ, Hama [65] Shibata M, Yamada S, Kumar SR, Calero M, Bading J, Fran-
E, Sekine-Aizawa Y, Saido TC (2000) Identification of gione B, Holtzman DM, Miller CA, Strickland DK, Ghiso J,
the major Abeta1-42-degrading catabolic pathway in brain Zlokovic BV (2000) Clearance of Alzheimer’s amyloid-ss(1-
parenchyma: suppression leads to biochemical and patho- 40) peptide from brain by LDL receptor-related protein-1 at
logical deposition. Nat Med 6, 143-150. the blood-brain barrier. J Clin Invest 106, 1489-1499.
[52] Iwata N, Tsubuki S, Takaki Y, Shirotani K, Lu B, Gerard [66] Deane R, Du Yan S, Submamaryan RK, LaRue B, Jovanovic
NP, Gerard C, Hama E, Lee HJ, Saido TC (2001) Metabolic S, Hogg E, Welch D, Manness L, Lin C, Yu J, Zhu H, Ghiso J,
regulation of brain Abeta by neprilysin. Science 292, 1550- Frangione B, Stern A, Schmidt AM, Armstrong DL, Arnold
1552. B, Liliensiek B, Nawroth P, Hofman F, Kindy M, Stern D,
[53] Marr RA, Rockenstein E, Mukherjee A, Kindy MS, Hersh Zlokovic B (2003) RAGE mediates amyloid-beta peptide
LB, Gage FH, Verma IM, Masliah E (2003) Neprilysin gene transport across the blood-brain barrier and accumulation in
transfer reduces human amyloid pathology in transgenic brain. Nat Med 9, 907-913.
mice. J Neurosci 23, 1992-1996. [67] DeMattos RB, Bales KR, Cummins DJ, Paul SM, Holtzman
DM (2002) Brain to plasma amyloid-beta efflux: a measure
M.P. Murphy and H. LeVine, III / AD Progression and the State of Aβ 321

of brain amyloid burden in a mouse model of Alzheimer’s S, Ausen B, Debnath ML, Barletta J, Price JC, Sandell J,
disease. Science 295, 2264-2267. Lopresti BJ, Wall A, Koivisto P, Antoni G, Mathis CA,
[68] Zlokovic BV (2005) Neurovascular mechanisms of Langstrom B (2004) Imaging brain amyloid in Alzheimer’s
Alzheimer’s neurodegeneration. Trends Neurosci 28, 202- disease with Pittsburgh Compound-B. Ann Neurol 55, 306-
208. 319.
[69] Roher AE, Chaney MO, Kuo YM, Webster SD, Stine WB, [83] Grimmer T, Henriksen G, Wester HJ, Forstl H, Klunk WE,
Haverkamp LJ, Woods AS, Cotter RJ, Tuohy JM, Krafft GA, Mathis CA, Kurz A, Drzezga A (2008) Clinical severity of
Bonnell BS, Emmerling MR (1996) Morphology and toxicity Alzheimer’s disease is associated with PIB uptake in PET.
of Abeta-(1-42) dimer derived from neuritic and vascular Neurobiol Aging, in press.
amyloid deposits of Alzheimer’s disease. J Biol Chem 271, [84] Klunk WE, Lopresti BJ, Ikonomovic MD, Lefterov IM,
20631-20635. Koldamova RP, Abrahamson EE, Debnath ML, Holt DP,
[70] Roher A, Wolfe D, Palutke M, KuKuruga D (1986) Purifi- Huang GF, Shao L, DeKosky ST, Price JC, Mathis CA (2005)
cation, ultrastructure, and chemical analysis of Alzheimer Binding of the positron emission tomography tracer Pitts-
disease amyloid plaque core protein. Proc Natl Acad Sci U S burgh compound-B reflects the amount of amyloid-beta in
A 83, 2662-2666. Alzheimer’s disease brain but not in transgenic mouse brain.
[71] Hasegawa M (2006) Biochemistry and molecular biology of J Neurosci 25, 10598-10606.
tauopathies. Neuropathology 26, 484-490. [85] Rosen RF, Walker LC, LeVine H, 3rd (2009) PIB binding
[72] Steinerman JR, Irizarry M, Scarmeas N, Raju S, Brandt J, in aged primate brain: enrichment of high-affinity sites in
Albert M, Blacker D, Hyman B, Stern Y (2008) Distinct humans with Alzheimer’s disease. Neurobiol Aging, in press.
pools of beta-amyloid in Alzheimer disease-affected brain: [86] Maeda J, Ji B, Irie T, Tomiyama T, Maruyama M, Okauchi T,
a clinicopathologic study. Arch Neurol 65, 906-912. Staufenbiel M, Iwata N, Ono M, Saido TC, Suzuki K, Mori
[73] Murphy MP, Beckett TL, Ding Q, Patel E, Markesbery WR, H, Higuchi M, Suhara T (2007) Longitudinal, quantitative
St Clair DK, LeVine H, 3rd, Keller JN (2007) Abeta solubil- assessment of amyloid, neuroinflammation, and anti-amyloid
ity and deposition during AD progression and in APPxPS-1 treatment in a living mouse model of Alzheimer’s disease
knock-in mice. Neurobiol Dis 27, 301-311. enabled by positron emission tomography. J Neurosci 27,
[74] Dickson TC, Vickers JC (2001) The morphological pheno- 10957-10968.
type of beta-amyloid plaques and associated neuritic changes [87] Meyer-Luehmann M, Coomaraswamy J, Bolmont T, Kaeser
in Alzheimer’s disease. Neuroscience 105, 99-107. S, Schaefer C, Kilger E, Neuenschwander A, Abramowski D,
[75] O’Nuallain B, Wetzel R (2002) Conformational Abs recog- Frey P, Jaton AL, Vigouret JM, Paganetti P, Walsh DM, Math-
nizing a generic amyloid fibril epitope. Proc Natl Acad Sci ews PM, Ghiso J, Staufenbiel M, Walker LC, Jucker M (2006)
U S A 99, 1485-1490. Exogenous induction of cerebral beta-amyloidogenesis is
[76] Lambert MP, Velasco PT, Chang L, Viola KL, Fernandez S, governed by agent and host. Science 313, 1781-1784.
Lacor PN, Khuon D, Gong Y, Bigio EH, Shaw P, De Felice [88] Kane MD, Lipinski WJ, Callahan MJ, Bian F, Durham
FG, Krafft GA, Klein WL (2007) Monoclonal antibodies that RA, Schwarz RD, Roher AE, Walker LC (2000) Evidence
target pathological assemblies of Abeta. J Neurochem 100, for seeding of beta -amyloid by intracerebral infusion of
23-35. Alzheimer brain extracts in beta -amyloid precursor protein-
[77] Nilsson KP, Aslund A, Berg I, Nystrom S, Konradsson P, transgenic mice. J Neurosci 20, 3606-3611.
Herland A, Inganas O, Stabo-Eeg F, Lindgren M, Wester- [89] Walker LC, Bian F, Callahan MJ, Lipinski WJ, Durham RA,
mark GT, Lannfelt L, Nilsson LN, Hammarstrom P (2007) LeVine H (2002) Modeling Alzheimer’s disease and other
Imaging distinct conformational states of amyloid-beta fib- proteopathies in vivo: is seeding the key? Amino Acids 23,
rils in Alzheimer’s disease using novel luminescent probes. 87-93.
ACS Chem Biol 2, 553-560. [90] Deshpande A, Mina E, Glabe C, Busciglio J (2006) Differ-
[78] Petkova AT, Leapman RD, Guo Z, Yau WM, Mattson MP, ent conformations of amyloid beta induce neurotoxicity by
Tycko R (2005) Self-propagating, molecular-level polymor- distinct mechanisms in human cortical neurons. J Neurosci
phism in Alzheimer’s beta-amyloid fibrils. Science 307, 262- 26, 6011-6018.
265. [91] Koffie RM, Meyer-Luehmann M, Hashimoto T, Adams KW,
[79] Ray S, Britschgi M, Herbert C, Takeda-Uchimura Y, Boxer Mielke ML, Garcia-Alloza M, Micheva KD, Smith SJ, Kim
A, Blennow K, Friedman LF, Galasko DR, Jutel M, Karydas ML, Lee VM, Hyman BT, Spires-Jones TL (2009) Oligomer-
A, Kaye JA, Leszek J, Miller BL, Minthon L, Quinn JF, ic amyloid {beta} associates with postsynaptic densities and
Rabinovici GD, Robinson WH, Sabbagh MN, So YT, Sparks correlates with excitatory synapse loss near senile plaques.
DL, Tabaton M, Tinklenberg J, Yesavage JA, Tibshirani R, Proc Natl Acad Sci U S A 106, 4012-4017.
Wyss-Coray T (2007) Classification and prediction of clinical [92] Bitan G, Kirkitadze MD, Lomakin A, Vollers SS, Benedek
Alzheimer’s diagnosis based on plasma signaling proteins. GB, Teplow DB (2003) Amyloid beta -protein (Abeta) as-
Nat Med 13, 1359-1362. sembly: Abeta 40 and Abeta 42 oligomerize through distinct
[80] Paravastu AK, Qahwash I, Leapman RD, Meredith SC, Ty- pathways. Proc Natl Acad Sci U S A 100, 330-335.
cko R (2009) Seeded growth of {beta}-amyloid fibrils from [93] Duering M, Grimm MO, Grimm HS, Schroder J, Hartmann
Alzheimer’s brain-derived fibrils produces a distinct fibril T (2005) Mean age of onset in familial Alzheimer’s disease
structure. Proc Natl Acad Sci U S A 106, 7443-7448. is determined by amyloid beta 42. Neurobiol Aging 26, 785-
[81] Wang Y, Klunk WE, Debnath ML, Huang GF, Holt DP, Shao 788.
L, Mathis CA (2004) Development of a PET/SPECT agent [94] Bitan G, Vollers SS, Teplow DB (2003) Elucidation of prima-
for amyloid imaging in Alzheimer’s disease. J Mol Neurosci ry structure elements controlling early amyloid beta-protein
24, 55-62. oligomerization. J Biol Chem 278, 34882-34889.
[82] Klunk WE, Engler H, Nordberg A, Wang Y, Blomqvist G, [95] Klyubin I, Betts V, Welzel AT, Blennow K, Zetterberg H,
Holt DP, Bergstrom M, Savitcheva I, Huang GF, Estrada Wallin A, Lemere CA, Cullen WK, Peng Y, Wisniewski T,
322 M.P. Murphy and H. LeVine, III / AD Progression and the State of Aβ

Selkoe DJ, Anwyl R, Walsh DM, Rowan MJ (2008) Amyloid Pare JF, Duong TQ, Hopkins WD, Preuss TM, Walker LC
beta protein dimer-containing human CSF disrupts synaptic (2008) Tauopathy with paired helical filaments in an aged
plasticity: prevention by systemic passive immunization. J chimpanzee. J Comp Neurol 509, 259-270.
Neurosci 28, 4231-4237. [111] Satou T, Cummings BJ, Head E, Nielson KA, Hahn FF, Mil-
[96] Shankar GM, Li S, Mehta TH, Garcia-Munoz A, Shep- gram NW, Velazquez P, Cribbs DH, Tenner AJ, Cotman CW
ardson NE, Smith I, Brett FM, Farrell MA, Rowan MJ, (1997) The progression of beta-amyloid deposition in the
Lemere CA, Regan CM, Walsh DM, Sabatini BL, Selkoe DJ frontal cortex of the aged canine. Brain Res 774, 35-43.
(2008) Amyloid-beta protein dimers isolated directly from [112] Cummings BJ, Head E, Afagh AJ, Milgram NW, Cotman CW
Alzheimer’s brains impair synaptic plasticity and memory. (1996) Beta-amyloid accumulation correlates with cognitive
Nat Med 14, 837-842. dysfunction in the aged canine. Neurobiol Learning Memory
[97] Glabe CG (2008) Structural classification of toxic amyloid 66, 11-23.
oligomers. J Biol Chem 283, 29639-29643. [113] Cummings BJ, Satou T, Head E, Milgram NW, Cole GM,
[98] Kayed R, Sokolov Y, Edmonds B, McIntire TM, Milton SC, Savage MJ, Podlisny MB, Selkoe DJ, Siman R, Greenberg
Hall JE, Glabe CG (2004) Permeabilization of lipid bilay- BD, Cotman CW (1996) Diffuse plaques contain C-terminal
ers is a common conformation-dependent activity of solu- A beta 42 and not A beta 40: evidence from cats and dogs.
ble amyloid oligomers in protein misfolding diseases. J Biol Neurobiol Aging 17, 653-659.
Chem 279, 46363-46366. [114] Head E, Pop V, Vasilevko V, Hill M, Saing T, Sarsoza F,
[99] Sokolov Y, Kozak JA, Kayed R, Chanturiya A, Glabe C, Nistor M, Christie LA, Milton S, Glabe C, Barrett E, Cribbs D
Hall JE (2006) Soluble amyloid oligomers increase bilayer (2008) A two-year study with fibrillar beta-amyloid (Abeta)
conductance by altering dielectric structure. J Gen Physiol immunization in aged canines: effects on cognitive function
128, 637-647. and brain Abeta. J Neurosci 28, 3555-3566.
[100] Georganopoulou DG, Chang L, Nam JM, Thaxton CS, Muf- [115] Games D, Buttini M, Kobayashi D, Schenk D, Seubert
son EJ, Klein WL, Mirkin CA (2005) Nanoparticle-based P (2006) Mice as models: transgenic approaches and
detection in cerebral spinal fluid of a soluble pathogenic Alzheimer’s disease. J Alzheimers Dis 9, 133-149.
biomarker for Alzheimer’s disease. Proc Natl Acad Sci U S [116] McGowan E, Eriksen J, Hutton M (2006) A decade of mod-
A 102, 2273-2276. eling Alzheimer’s disease in transgenic mice. Trends Genet
[101] Necula M, Kayed R, Milton S, Glabe CG (2007) Small 22, 281-289.
molecule inhibitors of aggregation indicate that amyloid beta [117] LeVine H, Walker LC (2006) Model’s of Alzheimer’s Disease
oligomerization and fibrillization pathways are independent in: Handbook of Models for Human Aging, Conn MP, ed.,
and distinct. J Biol Chem 282, 10311-10324. Academic Press/Elsevier, New York, pp. 121-134.
[102] Esler WP, Stimson ER, Jennings JM, Vinters HV, Ghilardi [118] Oakley H, Cole SL, Logan S, Maus E, Shao P, Craft J,
JR, Lee JP, Mantyh PW, Maggio JE (2000) Alzheimer’s dis- Guillozet-Bongaarts A, Ohno M, Disterhoft J, Van Eldik L,
ease amyloid propagation by a template-dependent dock-lock Berry R, Vassar R (2006) Intraneuronal beta-amyloid aggre-
mechanism. Biochemistry 39, 6288-6295. gates, neurodegeneration, and neuron loss in transgenic mice
[103] O’Nuallain B, Williams AD, Westermark P, Wetzel R (2004) with five familial Alzheimer’s disease mutations: potential
Seeding specificity in amyloid growth induced by heterolo- factors in amyloid plaque formation. J Neurosci 26, 10129-
gous fibrils. J Biol Chem 279, 17490-17499. 10140.
[104] Meyer-Luehmann M, Spires-Jones TL, Prada C, Garcia- [119] Flood DG, Reaume AG, Dorfman KS, Lin YG, Lang DM,
Alloza M, de Calignon A, Rozkalne A, Koenigsknecht- Trusko SP, Savage MJ, Annaert WG, De Strooper B, Siman
Talboo J, Holtzman DM, Bacskai BJ, Hyman BT (2008) R, Scott RW (2002) FAD mutant PS-1 gene-targeted mice:
Rapid appearance and local toxicity of amyloid-beta plaques increased A beta 42 and A beta deposition without APP
in a mouse model of Alzheimer’s disease. Nature 451, 720- overproduction. Neurobiol Aging 23, 335-348.
724. [120] Fung J, Frost D, Chakrabartty A, McLaurin J (2004) Interac-
[105] Robbins EM, Betensky RA, Domnitz SB, Purcell SM, tion of human and mouse Abeta peptides. J Neurochem 91,
Garcia-Alloza M, Greenberg C, Rebeck GW, Hyman BT, 1398-1403.
Greenberg SM, Frosch MP, Bacskai BJ (2006) Kinetics [121] Jankowsky JL, Younkin LH, Gonzales V, Fadale DJ, Slunt
of cerebral amyloid angiopathy progression in a transgenic HH, Lester HA, Younkin SG, Borchelt DR (2007) Rodent
mouse model of Alzheimer disease. J Neurosci 26, 365-371. A beta modulates the solubility and distribution of amyloid
[106] Lemere CA, Maier M, Jiang L, Peng Y, Seabrook TJ (2006) deposits in transgenic mice. J Biol Chem 282, 22707-22720.
Amyloid-beta immunotherapy for the prevention and treat- [122] Ye L, Morgenstern JL, Lamb JR, Lockhart A (2006) Charac-
ment of Alzheimer disease: lessons from mice, monkeys, terisation of the binding of amyloid imaging tracers to rodent
and humans. Rejuvenation Res 9, 77-84. Abeta fibrils and rodent-human Abeta co-polymers. Biochem
[107] Wisniewski HM, Ghetti B, Terry RD (1973) Neuritic (senile) Biophys Res Commun 347, 669-677.
plaques and filamentous changes in aged rhesus monkeys. J [123] McGowan E, Pickford F, Kim J, Onstead L, Eriksen J, Yu C,
Neuropathol Exp Neurol 32, 566-584. Skipper L, Murphy MP, Beard J, Das P, Jansen K, Delucia
[108] Walker LC, Kitt CA, Schwam E, Buckwald B, Garcia F, M, Lin WL, Dolios G, Wang R, Eckman CB, Dickson DW,
Sepinwall J, Price DL (1987) Senile plaques in aged squirrel Hutton M, Hardy J, Golde T (2005) Abeta42 is essential
monkeys. Neurobiol Aging 8, 291-296. for parenchymal and vascular amyloid deposition in mice.
[109] Martin LJ, Sisodia SS, Koo EH, Cork LC, Dellovade TL, Neuron 47, 191-199.
Weidemann A, Beyreuther K, Masters C, Price DL (1991) [124] Gotz J, Chen F, van Dorpe J, Nitsch RM (2001) Formation of
Amyloid precursor protein in aged nonhuman primates. Proc neurofibrillary tangles in P301l tau transgenic mice induced
Natl Acad Sci U S A 88, 1461-1465. by Abeta 42 fibrils. Science 293, 1491-1495.
[110] Rosen RF, Farberg AS, Gearing M, Dooyema J, Long PM, [125] Lewis J, Dickson DW, Lin WL, Chisholm L, Corral A, Jones
Anderson DC, Davis-Turak J, Coppola G, Geschwind DH, G, Yen SH, Sahara N, Skipper L, Yager D, Eckman C, Hardy
M.P. Murphy and H. LeVine, III / AD Progression and the State of Aβ 323

J, Hutton M, McGowan E (2001) Enhanced neurofibrillary the chemically modified and insoluble peptides deposited in
degeneration in transgenic mice expressing mutant tau and Alzheimer’s disease senile plaques. Biochemistry 41, 922-
APP. Science 293, 1487-1491. 928.
[126] Walsh DM, Klyubin I, Fadeeva JV, Cullen WK, Anwyl R, [129] Ye L, Morgenstern JL, Gee AD, Hong G, Brown J, Lock-
Wolfe MS, Rowan MJ, Selkoe DJ (2002) Naturally secreted hart A (2005) Delineation of positron emission tomography
oligomers of amyloid beta protein potently inhibit hippocam- imaging agent binding sites on beta-amyloid peptide fibrils.
pal long-term potentiation in vivo. Nature 416, 535-539. J Biol Chem 280, 23599-23604.
[127] Kayed R, Head E, Thompson JL, McIntire TM, Milton SC, [130] LeVine H, 3rd (2005) Multiple ligand binding sites on A
Cotman CW, Glabe CG (2003) Common structure of soluble beta(1-40) fibrils. Amyloid 12, 5-14.
amyloid oligomers implies common mechanism of patho- [131] Lockhart A, Ye L, Judd DB, Merritt AT, Lowe PN, Morgen-
genesis. Science 300, 486-489. stern JL, Hong G, Gee AD, Brown J (2005) Evidence for
[128] Kalback W, Watson MD, Kokjohn TA, Kuo YM, Weiss the presence of three distinct binding sites for the thioflavin
N, Luehrs DC, Lopez J, Brune D, Sisodia SS, Staufenbiel T class of Alzheimer’s disease PET imaging agents on beta-
M, Emmerling M, Roher AE (2002) APP transgenic mice amyloid peptide fibrils. J Biol Chem 280, 7677-7684.
Tg2576 accumulate Abeta peptides that are distinct from

You might also like