11
Freund & Pettman, U.K. Reviews in the Neurosciences 21, 83-93 (2010) VOLUME 21, NO. 1, 2010 67 The monomer state of beta-amyloid: where the Alzheimer’s disease protein meets physiology M.L. Giuffrida 1,2 , F. Caraci 2 , P. De Bona 3 , G. Pappalardo 4 , F. Nicoletti 5,6 , E. Rizzarelli 3 , A. Copani 2,4 1 I.N.B.B. Fellowship, 2 Department of Pharmaceutical Sciences, 3 Department of Chemical Sciences, University of Catania, and 4 Institute of Biostructure and Bioimaging, National Research Council, Viale Andrea Doria, Catania 95125, Italy; 5 Department of Human Physiology and Pharmacology, University of Rome “La Sapienza”, Piazzale Aldo Moro, Rome 00185, Italy; 6 Instituto Neurologico Mediterraneo, Neuromed, Località Camerelle, Pozzilli 86077, Italy SYNOPSIS One hundred years of study have identified beta-Amyloid (Aβ) as the most interesting feature of Alzheimer’s disease (AD). Since the discovery of Aβ as the principal component of amyloid plaques, the central challenge in AD research has been the understanding of Aβ involvement in the neurodegenerative process of the disease. The ability of Aβ to undergo conformational changes and subsequent aggre- gation has always been a limiting factor in finding out the activities of the peptide. Extensive research has been carried out to study the molecular mechanisms of amyloid self-assembly. The finding that soluble Aβ concentrations in the brain are correlated with the severity of AD, whereas fibrillar density is not /40,42/, has pointed attention toward the oligomeric forms of Aβ, which are generally considered the most toxic and, therefore, the most important species to be addressed. Despite great efforts in basic AD research, none of the currently available treatments is able to treat the devastating effects of the disease, leading to the consideration that there is more to reason _____________________________ Accepted: February 10, 2010 Address for correspondence: A. Copani Department of Pharmaceutical Sciences University of Catania, Viale Andrea Doria 6, Catania 95125, Italy; phone: +39-095-7384212. e-mail: [email protected] than just Aβ production and aggregation. Here we summarize the emerging evidence for the physiological functions of Aβ, including our recent demonstration that Aβ monomers are endowed with neuroprotective activity, and propose that Aß aggregation might contribute to AD pathology through a “loss-of-function” process. Finally, we discuss the current therapeutics targeting the cerebral load of Aβ and possible new ones aimed at preserving the biological functions of Aβ. KEYWORDS Aβ monomers, insulin/IGF-1 receptor, excitotoxicity, neuroprotection, aggregation inhibitors SOLUBLE AΒ OLIGOMERS AS INITIATING FACTORS IN ALZHEIMER’S DISEASE In the controversial literature about Alzheimer’s disease (AD), a predominant idea that seems to be universally accepted refers to the crucial role of Aβ in the pathogenesis of the disease. The “amyloid cascade hypothesis” can be considered the first effort in combining and harmonizing the great amount of findings on Aβ in AD /25/. The hypothesis summarizes the sequential steps occurring during the aggregation process of Aβ, with the earliest event of the cascade, being the increased production of Aβ monomers that arise either from missense mutations in specific genes linked to the familial form of the disease or from multifactor causes in

The Monomer State of Beta-Amyloid: Where the Alzheimer's Disease Protein Meets Physiology

  • Upload
    cnr-it

  • View
    0

  • Download
    0

Embed Size (px)

Citation preview

Freund & Pettman, U.K. Reviews in the Neurosciences 21, 83-93 (2010)

VOLUME 21, NO. 1, 2010 67

The monomer state of beta-amyloid: where the Alzheimer’s

disease protein meets physiology

M.L. Giuffrida1,2

, F. Caraci2, P. De Bona

3, G. Pappalardo

4, F. Nicoletti

5,6, E. Rizzarelli

3, A. Copani

2,4

1I.N.B.B. Fellowship,

2Department of Pharmaceutical Sciences,

3Department of Chemical Sciences,

University of Catania, and 4Institute of Biostructure and Bioimaging, National Research Council,

Viale Andrea Doria, Catania 95125, Italy; 5Department of Human Physiology and Pharmacology,

University of Rome “La Sapienza”, Piazzale Aldo Moro, Rome 00185, Italy; 6Instituto Neurologico

Mediterraneo, Neuromed, Località Camerelle, Pozzilli 86077, Italy

SYNOPSIS

One hundred years of study have identified

beta-Amyloid (Aβ) as the most interesting

feature of Alzheimer’s disease (AD). Since the

discovery of Aβ as the principal component of

amyloid plaques, the central challenge in AD

research has been the understanding of Aβ

involvement in the neurodegenerative process of

the disease. The ability of Aβ to undergo

conformational changes and subsequent aggre-

gation has always been a limiting factor in

finding out the activities of the peptide.

Extensive research has been carried out to

study the molecular mechanisms of amyloid

self-assembly. The finding that soluble Aβ

concentrations in the brain are correlated with

the severity of AD, whereas fibrillar density is

not /40,42/, has pointed attention toward the

oligomeric forms of Aβ, which are generally

considered the most toxic and, therefore, the

most important species to be addressed. Despite

great efforts in basic AD research, none of the

currently available treatments is able to treat

the devastating effects of the disease, leading to

the consideration that there is more to reason

_____________________________ Accepted: February 10, 2010

Address for correspondence:

A. Copani

Department of Pharmaceutical Sciences

University of Catania, Viale Andrea Doria 6, Catania

95125, Italy; phone: +39-095-7384212.

e-mail: [email protected]

than just Aβ production and aggregation. Here

we summarize the emerging evidence for the

physiological functions of Aβ, including our

recent demonstration that Aβ monomers are

endowed with neuroprotective activity, and

propose that Aß aggregation might contribute

to AD pathology through a “loss-of-function”

process. Finally, we discuss the current

therapeutics targeting the cerebral load of Aβ

and possible new ones aimed at preserving the

biological functions of Aβ.

KEYWORDS

Aβ monomers, insulin/IGF-1 receptor, excitotoxicity,

neuroprotection, aggregation inhibitors

SOLUBLE AΒ OLIGOMERS AS INITIATING

FACTORS IN ALZHEIMER’S DISEASE

In the controversial literature about

Alzheimer’s disease (AD), a predominant idea that

seems to be universally accepted refers to the

crucial role of Aβ in the pathogenesis of the

disease. The “amyloid cascade hypothesis” can be

considered the first effort in combining and

harmonizing the great amount of findings on Aβ in

AD /25/. The hypothesis summarizes the

sequential steps occurring during the aggregation

process of Aβ, with the earliest event of the

cascade, being the increased production of Aβ

monomers that arise either from missense

mutations in specific genes linked to the familial

form of the disease or from multifactor causes in

M.L. GIUFFRIDA ET AL.

REVIEWS IN THE NEUROSCIENCES

84

the sporadic form of AD. The change of Aβ

folding is another crucial event that influences the

aggregation properties of the peptide, and gives

rise to the formation of soluble Aβ aggregates (i.e.,

oligomers). Soluble Aβ oligomers correlate better

with dementia than insoluble fibrillar deposits /40/,

suggesting that peptide oligomers may represent

the primary neurotoxic species in AD. The

neurotoxicity of Aβ oligomers has been confirmed

by distinct experimental approaches, including the

use of synthetic or native Aβ peptides, cell culture

systems over-expressing the amyloid precursor

protein (APP) from which Aβ is derived, and APP

transgenic mice /75,38/.

Today, diffusible oligomers of Aβ are

considered metastable neurotoxic molecules that

exist for prolonged periods without conversion to

fibrillar structures. Therefore, attention has been

focused on Aβ oligomeric species to elucidate the

underlying mechanism of neuronal degeneration.

Many different types of assembly forms of

synthetic Aβ, including protofibrils (PFs), annular

structures, paranuclei, Aβ-derived diffusible

ligands (ADDL), and globulomers have been

described over the last two decades /71/. In

particular, ADDLs have been shown to inhibit

hippocampal long-term potentiation /77/ and to

cause death in different culture systems /33,29/.

The pathogenic relevance of natural Aβ

oligomers is supported by the finding that their

formation is increased by expressing AD-causing

mutations within APP or presenilin genes in

recombinant cells /83/. Moreover, putative ADDL-

like oligomeric assemblies have been isolated from

post-mortem AD brains and their presence

correlates with memory loss /21/. Natural

oligomers of human Aβ are acutely toxic on

synaptic functions when micro-injected in living

rats /75/ or added in vitro to hippocampal slices

/73/. In rats, Aβ oligomers have also been shown

to interfere rapidly and reversibly with the memory

of a learned behavior /30/. Noteworthy, the

evidence that Aβ immunotherapy neutralizes the

synapto-toxic effects of soluble oligomers /30/ has

led to the notion that antibody-mediated

inactivation of Aβ oligomers might be a

therapeutic strategy for early AD /60/.

AΒ AS A MODULATOR OF SYNAPTIC

ACTIVITY: THE UNKNOWN SPECIES

The production of the Aβ through the endo-

proteolytic cleavage of APP is a physiological

process that occurs normally in neuronal cells /8/.

After production, Aβ is exported outside the brain

by the low density lipoprotein receptor related

protein-1 (LRP-1). The amount of Aβ synthesized

outside of the brain is instead transported inside via

the receptor for advanced glycation end-products

/41/. The tightly regulated bidirectional trafficking

of Aβ across the blood brain barrier suggests a

biological role of the protein for which the

production and removal of the peptide must be

maintained into a specific range of concentrations.

Accordingly, several metalloproteases, including

neprilysin, insulin-degrading enzyme (IDE) and

endothelin converting enzymes, have been reported

to act in Aβ clearance /16,58/.

During the past decade, few physiological

activities have been proposed for the peptide. More

recently, the use of transgenic mice has provided a

further hint toward this concept. β-Amyloid

precursor protein cleavage enzyme (BACE 1)

knock-out mice, which lack Aβ formation, have

behavioral deficits /26/ and synaptic dysfunctions

/49,76/, including a reduced activity-dependent

strengthening of presynaptic release at mossy fiber

synapses /76/.

Similar to BACE 1 knock-out, APP null

mutant mice show an impaired formation of LTP

and, as a consequence of this synaptic impairment,

they have reduced learning and memory /45,10,

64/. These finding might well be related to the loss

of function of either BACE 1 or APP, rather than

to the missing production of Aβ. Interestingly

enough, however, the transgenic approach

strengths the older finding that physiological Aβ

production sustains survival in cultured neurons

/56/. Along this line is the demonstration that

picomolar concentrations of synthetic Aβ, which

are likely to approximate the endogenous level of

the peptide, enhance synaptic plasticity and

memory in the hippocampus /57/. In the same

system, high nanomolar concentrations of Aβ led

to the well known impairment of synaptic

functions /57/, suggesting that the concentration

MONOMER STATE OF BETA-AMYLOID

VOLUME 21, NO. 1, 2010

85

level of the peptide is crucial for its physiological

activity.

Several lines of evidence converge to indicate

that Aβ is released in normal brains during

synaptic activity. Kamenetz and colleagues /28/

first reported that Aβ is secreted from healthy

neurons in response to neuronal activity, and in

turn, can down-regulate excitatory synaptic

transmission. This negative feedback loop, in

which neuronal activity promotes Aβ production

and Aβ decreases synaptic activity, would provide

a physiological homeostatic mechanism to

maintain the levels of neuronal activity. Recently,

the regulation of the endogenous synaptic release

of Aβ has been addressed in rodent hippocampal

cells and slices /1/. This study shows that acute

increases in Aβ levels expand reversibly the

number of active synapses and the amount of

neurotransmitter released at each synapse, whereas

enduring inhibition of Aβ clearance results in a

reduction in the number of synapses. Thus, Aβ

appears to be a modulator of synaptic activity

requiring a fine balance between production and

removal. Accordingly, sequestration of endogenous

Aβ by the monoclonal antibody 4G8 disrupts

memory in adult rats, whereas hippocampal

injection of physiological concentrations of Aβ

rescues the amnesia produced by the anti-Aβ

antibody /19/.

Different from normal Aβ concentrations, the

high levels of peptide present in transgenic mice

over-expressing human APP are per se sufficient

to elicit epileptiform activity and seizure, even at

an early stage of the pathology and in the absence

of neuronal loss /52/. This Aβ–induced aberrant

neuronal activity has been suggested to trigger

compensatory inhibitory responses causally linked

to cognitive decline. In AD patients, 7% to 21% of

individuals with sporadic AD are estimated to have

at least one unprovoked clinically apparent seizure

during the illness. The relationship between this

phenomenon and AD is even stronger in the case

of autosomal dominant early onset AD /51/.

Once again, the levels of soluble Aβ may be

critical for the dual effect of Aβ at the synapse.

The discovery that Aβ binds to the 7 subunit of

nicotinic acetylcholine receptors (nAChRs) with

high affinity has provided strong support to the old

hypothesis of a cholinergic deficit responsible for

the cognitive dysfunction in AD (reviewed in /48/.

Nevertheless, whereas higher concentrations of Aβ

desensitize 7-containing nAChRs /55,35/, low

concentrations of the peptide appear to activate

pre-synaptic nAChRs /13,35/, which are responsible

for glutamate release during LTP.

Additional effects of Aβ at the synapse have

been reported as being solely disruptive and linked

to an impairment of AMPA and NMDA receptor

trafficking /67,23/, or to the disassembly of the

post-synaptic density /59/ Aβ. These and similar

studies refer to synthetic preparations of Aβ

oligomers, and clearly suggest that the concen-

trations of Aβ in the synaptic cleft affect its

aggregation state, and that differently assembled

Aβ aggregates have different effects on synaptic

activity. Nevertheless, the true identity of the Aβ

species that act as modulators of synaptic activity,

especially in the case of endogenous released Aβ

/1/ remains unclear.

AΒ MONOMER: THE NEUROPROTECTIVE

SPECIES

Based on the notion that synaptic activity

regulates the expression of gene products that are

important for neuronal survival /78/, the evidence

that Aβ can act as a synaptic modulator is per se

suggestive of a pro survival role of the peptide. One

of the first pieces of evidence supporting a

physiological role for Aβ dates back to 1989, when

the 1-28 fragment of the peptide was shown to have

neurotrophic activity /79/. The following years have

been characterized, instead, by an extensive amount

of research on the toxic effects of aggregated forms

of Aβ. Recently, emerging interest in the putative

physiological roles of Aβ has provided new

interesting data. Indirect evidence for the

implication of Aβ in the normal neuronal

metabolism can be found in several papers

published in this field. The in vitro inhibition of

either β- or γ-secretase seems to affect the viability

of cortical neurons, which are rescued by adding

picomolar concentrations of Aβ1-40/42 /56/.

The contribution of Aβ to physiological

neuronal activity is strengthened by the

observation that the addition of Aβ(1-42) to

cultured neurons enhances glucose uptake and

M.L. GIUFFRIDA ET AL.

REVIEWS IN THE NEUROSCIENCES

86

metabolism via the induction of hypoxia-inducible

factor-1 /69/. More important, indirect evidence for

a neuroprotective activity of Aβ has been recently

obtained in patients who underwent invasive

intracranial monitoring after acute brain injury.

The results provided by the authors show a strong

correlation between the Aβ levels in the cerebral

interstitial fluid and the patients’ neurological

status, with Aβ concentrations increasing when the

neurological status improves and falling when the

neurological status declines. /6/.

We recently identified the nature of the neuro-

protective effect of Aβ1-40/42, demonstrating that

the protective activity of Aβ is confined to

monomeric, low concentrated form of the peptide.

In neurons undergoing death by trophic deprivation,

synthetic Aβ(1-42) monomers had a rescuing effect

mediated by the activation of the phosphatidyl-

inositol-3-kinase (PI-3-K) pathway. The activation

of the PI-3-K pathway, which is a main surviving

path in neurons /18/, could be reconducted to the

stimulation of IGF-1 receptors and/or other

receptors of the insulin superfamily /20/.

Interestingly, Aβ1-40/42 monomers had a broad

rescuing effect that included neuroprotection against

excitotoxic cell death, a process that contributes to

several neurodegenerative diseases /14/.

A dysregulation of Insulin/IGF-1 signaling is

thought to sustain a crucial role in the pathogenesis

of AD. Some evidence indicates that insulin/IGF-1

resistance, as occurs in type 2 diabetes, is linked to

the development of late-onset forms of AD /39,61/

and alterations of both insulin receptors and IGF-1

receptors have been reported in the AD brain

/44,12/.

Zhao and co-workers have suggested that

insulin resistance in the AD brain is a response to

Aβ oligomers (ADDLs), which downregulate

neuronal surface insulin receptors /85/. On the

contrary, insulin/IGF-1 receptor activation seems

to promote the reduction of ADDLs to monomers

via the insulin-degrading enzyme (IDE) activity

/84/. According to this evidence, accumulating

toxic Aβ species impair insulin/IGF-1 signaling

that in turn will exacerbate Aβ aggregation with

ensuing neurotoxicity. Interestingly, the other way

around would be that Aβ monomers sustain

insulin/IGF-1 signaling that in turn will impede

oligomerization (Fig. 1).

We should highlight that monomers of the

arctic-mutant Aβ(1-42) do not share the same

neuroprotective properties of the Aβ40/42 peptides

/20/. Conformational studies of the different Aβ

monomers indicate that the neuroprotective Aβ40/

42 species share similar folding properties and

have similar conformational features /20/, thus

suggesting that they might bind to specific

recognition sites on the neuronal surface. Whether

Aβ40/42 bind directly to IGF1/insulin receptors

remains to be established.

CURRENT STATUS OF DISEASE-MODYFING

DRUGS IN AD AND THE POTENTIAL ROLE OF

A MONOMERS FOR DESIGNING NEW

PHARMACOLOGICAL STRATEGIES

In the process leading to oligomers formation

from APP production, many steps could potentially

be targeted for the treatment of AD. Immuno-

therapy can be considered an approach potentially

able to target the production, aggregation, and

deposition of Aβ, and anti-Aβ therapy is

considered one of the most interesting possibilities

for the development of disease-modifying drugs in

AD /36,72/. Active immunization promotes the

formation of antibodies against Aβ by stimulating

an immune response in the patient, whereas

passive immunotherapy supplies antibodies from

an exogenous source /72/. Active Aβ immuno-

therapy has been studied and validated since 1999

in AD mouse models, in which the generation of

Aβ antibodies results in the clearance of cerebral

amyloid plaques, a process dependent on

microglial phagocytosis of antibody-opsonized Aβ

deposits /63/. Aβ immunotherapy improves

cognitive deficits in AD mouse models and lowers

the plaque load in non-human primates /36/.

Unfortunately, a Phase II clinical trial of active

immunization (the AN-1792 vaccine), using full-

length human Aβ(1-42) peptide with QS-21

adjuvant, had to be stopped prematurely in 2002

because approximately 6% of patients developed

aseptic meningoencephalitis /7/. Different

mechanisms have been proposed to explain the

negative side effects of the AN-1792 vaccine trial,

including the occurrence of a T-cell recognition of

the human full-length Aβ as a self-protein, which

MONOMER STATE OF BETA-AMYLOID

VOLUME 21, NO. 1, 2010

87

Fig 1: Schematic drawing of the interactions between Aβ(1-42) and the insulin/IGF-1 receptor signaling (IR/IGF-1R).

Monomeric forms of Aβ promote the activation of the insulin/IGF-1 signaling, resulting into a sustained

neuronal survival (via the activation of the PI-3K pathway) and self-maintened levels of Aβ monomers (via the

activity of the insulin-degrading enzyme -IDE- ). On the contrary, accumulating ADDLs induce the

downregulation of insulin/IGF-1 receptors that will exacerbate Aβ aggregation with ensuing neurotoxicity.

Dashed lines refer to uncharecterized mechanisms of action. Question mark refers to the unknown binding site

of Aβ monomers.

may have induced an adverse auto-immune response /81/. Data collected from patients over a period of 6 years following immunization with AN-1792 demonstrated that

the generation of anti-Aβ antibodies results in the

clearance of amyloid plaques in the AD brain, but

this clearance does not prevent progressive neuro-

degeneration /46,27/. Thus, it seems that removing

fibrillar amyloid from the brain may not be

sufficient to alter the course of AD. Furthermore,

significant aspects of AD pathology were

unaffected by vaccination, including the presence

of vascular amyloid and of hyper-phosphorylated

tau deposits /31/.

Alternative safer approaches for active

immunization are being developed, including those

using short Aβ immunogens that miss Aβ-specific

T cell epitopes (Aβ16-42) /36/.

Passive Aβ immunotherapy is being pursued

by administering monthly intravenous injections of

humanized Aβ monoclonal antibodies (i.e.,

bapineuzumab) to AD patients /62/. The results

from a phase II clinical trial indicate a potential

efficacy of bapineuzumab on cognitive functions in

APOE epsilon4 non-carriers with mild to moderate

AD, but not in APOE epsilon4 carriers (i.e.,

individuals carrying the major genetic risk factor

for AD), where this drug even favors the onset of a

vasogenic edema /62/.

Taking into consideration the role of Aβ

monomers in neuronal survival and, likely, in

synaptic plasticity and memory formation, anti-Aβ

M.L. GIUFFRIDA ET AL.

REVIEWS IN THE NEUROSCIENCES

88

antibodies should be designed to spare Aβ

monomers. At present, whether passive

immunotherapy with bapineuzumab may have

influenced Aβ monomer levels or functions is

unknown. Interestingly, it has been recently

demonstrated that free human Ig γ heavy chains

(HC), which ameliorate Aβ oligomer-induced

impairment of rodent hippocampal LTP, bind

selectively to fibrils and oligomers, but not to

native Aβ monomers /2/. Conformation-specific

antibodies, which bind with high specificity to

ADDLs, are being developed as a new

immunotherapeutic approach to AD and might

become available for clinical trials in the near

future /32/.

The best alternative to anti-Aβ immunotherapy

is believed to be the use of inhibitors of β-secretase

(BACE 1) and -secretase, due to the essential role

of these enzymes in the generation of Aβ from

APP /74/. Whereas only one BACE 1 inhibitor

(CTS-21166) has proceeded to clinical testing /53/,

-secretase inhibitors have failed to show a

significant clinical effect in trials conducted in

patients with mild to moderate AD /17/. Initial

positive results in mild AD were observed with

tarenflurbil, a selective Aβ-lowering agent (SALA)

/80/, but a very recent phase III 18-month,

randomized, placebo-controlled, double-blind trial

study clearly showed a non significant effect of

this drug in outcome measures of cognition /22/.

Overall, the anti-Aβ approaches in clinical

testing have been disappointing. The question now

is to understand why, so that future efforts can be

more successful. Indeed, as opposed to amyloid

plaques, Aβ oligomers were found increased in

some AD patients who received the AN-1792

vaccine /54/, suggesting that the elevated pool of

soluble Aβ oligomers was left unaltered or could

have promoted the progression of the disease. The

alternative explanation is that the vaccination

strategy might have removed the neuroprotective

Aβ monomers, thus exacerbating the course of the

disease. A depletion of Aβ monomers may explain

the lack of efficacy of tested -secretase inhibitors.

Low concentrations of Aβ(1-42) monomers, as

measured by different (enzyme-linked immuno-

absorbent assays) ELISAs, both in the CSF and in

the plasma, predict the conversion of mild

cognitive impairment (MCI) to AD, and parallel

brain Aβ deposition /37,82/. Similarly, low CSF

levels of Aβ(1-40), which is endowed with

neuroprotective activity as Aβ(1-42) /20/, have

been observed in MCI patients with a more rapid

cognitive decline /24/. More recently, low plasma

Aβ(1-42) levels have also been found in depressed

elders with cognitive deficits and no obvious risk

factor for AD (i.e., presence of ApoE4 allele) /70/.

To date, decreased levels of CSF or plasma Aβ(1-

42) monomers are considered a premorbid bio-

marker for AD that merely reflects the increasing

insolubility of Aβ in the brain, and does not

provide causality with AD. Nevertheless, given the

neuroprotective activity of Aβ monomers, we must

consider that a depletion of Aβ monomers in the

preclinical phase of AD might be involved in AD

progression.

Interestingly, reduced CSF Aβ levels are also

seen in neurodegenerative diseases other than AD,

including progressive supranuclear palsy and

corticobasal degeneration /47/ dementia with Lewy

bodies /43/, amyotrophic lateral sclerosis /66/, and

Creutzfeldt-Jacob disease /50/. Thus, decreased Aβ

levels in the CSF may occur without amyloid

deposition in the brain. Although the nature of this

event remains unclear, and probably relays on

disparate mechanisms affecting Aβ metabolism, it

strengthens the idea that Aß may functions as an

endogenous broad-spectrum neurotrophic factor.

A full comprehension of the mechanisms that

regulate A metabolism, together with the

elucidation of the factors that in vivo contribute to

maintain Aβ in an active monomeric conformation,

will be essential for the design a new generation of

therapeutics for AD. Along this line, the study of

metal dyshomeostasis related to A oligomeri-

zation in AD (reviewed in /65/) has led to the

development of metal-protein attenuating

compounds (MPACs) able to inhibit Zn2+

- and

Cu2+

-induced A oligomerizations without

affecting transition-metal homeostasis. PBT2 is the

only MPAC being tested in the clinic.

Interestingly, the results from a phase IIa trial with

PBT2 in 78 patients with early AD indicate a time-

and dose-dependent improvement of executive

functions over placebo in the PBT2 subject group

/34/. Recent findings /4/ indicated that copper and

zinc ionophores are able to induce the degradation

of A by the metal-dependent up-regulation of

MONOMER STATE OF BETA-AMYLOID

VOLUME 21, NO. 1, 2010

89

metalloproteases. It is intriguing that the

expression of metalloproteases by metal-

complexes depends on the activation of the PI-3-K

pathway, the downstream inhibition of the

glycogen synthase kinase 3and the activation of

extracellular regulated kinases /9/, suggesting that

metal-complexes might also have an impact on the

activation of neuroprotective signaling pathways.

The use of aggregation inhibitors can be

viewed as a promising strategy aimed at reducing

the transition of neuroprotective Aβ monomers into

toxic oligomeric species /9/. The notion that early,

soluble Aβ intermediates are key agents in the

cytotoxic effect causing neuronal death suggests

that a major effort should be directed toward the

inhibition of amyloid aggregation at very early

stages. Therefore, agents that target the basic

molecular recognition process preceding the

formation of early intermediates would be the most

valuable candidates. Either natural or synthesized

compounds are being studied, thus providing an

exciting future area for the development of new

therapeutics /3/. Current short peptides or peptide-

mimetics inhibitig A self-association /15/ act by

recognizing A amyloidogenic “hotspots” regions,

a process essentially driven by hydrophobic

interactions, and impeding further growth of the

well-ordered amyloid chain. Yet, peptidic anti-

fibrillogenic agents may be easily inactivated by

proteolysis.

We have conjugated a trehalose moiety to a

peptide inhibitor of A aggregation, the LPFFD,

also referred to as iA5p /68/, thus developing a

new class of protease-resistant inhibitors of A

toxicity /11/. According to the mechanism

proposed by Blackley et al. /5/, trehalose-

conjugated peptides do not necessarily interfere

with the elongation phase of the fibrils, but may act

instead at the level where the globular oligomer

species start to assemble. Interestingly, these novel

glyco-peptide inhibitors recognize and bind the

monomeric form of Athereby delaying and

reducing A aggregation burden (Fig. 2) /11/.

CONCLUSIONS

Originally considered a toxic waste of APP

metabolism, Aß is now revealed as an endogenous

regulator of synaptic activity and a neuroprotective

Fig 2: Schematic representation of the amyloid aggregation steps from monomeric to fibrillar assembly states. Unlike beta-sheet

breakers, glycopeptide inhibitors of Aβ aggregation act in a very early stage of the process as monomer stabilizers, halting

the formation of toxic intermediate species.

M.L. GIUFFRIDA ET AL.

REVIEWS IN THE NEUROSCIENCES

90

factor. This knowledge advances our understanding

of the processes leading to the neuropathology of

AD and conceivably will help us to design better

therapeutic strategies.

REFERENCES

1. Abramov E, Dolev I, Fogel H, Ciccotosto GD, Ruff

E, Slutsky I. Amyloid-beta as a positive endogenous

regulator of release probability at hippocampal

synapses. Nat Neurosci 2009;12: 1567-76.

2. Adekar SP, Klyubin I, Macy S, Rowan MJ,

Solomon A, Dessain SK, O'Nuallain B Inherent

anti-amyloidogenic activity of human Ig {gamma}

heavy chains. J Biol Chem 2010;285(2):1066-74.

3. Amijee H, Madine J, Middleton DA, Doig AJ.

Inhibitors of protein aggregation and toxicity.

Biochem Soc Trans 2009;37:692-6.

4. Bica L, Crouch PJ, Cappai R, White AR. Metallo-

complex activation of neuroprotective signalling

pathways as a therapeutic treatment for Alzheimer's

disease. Mol Biosyst 2009;5:134-42.

5. Blackley HK, Sanders GH, Davies MC, Roberts CJ,

Tendler SJ, Wilkinson MJ. In-situ atomic force

microscopy study of beta-amyloid fibrilli-zation. J

Mol Biol. 2000;298:833-40.

6. Brody DL, Magnoni S, Schwetye KE, Spinner ML,

Esparza TJ, Stocchetti N, Zipfel GJ, Holtzman DM.

Amyloid-beta dynamics correlate with neurological

status in the injured human brain. Science

2008;321:1221-4.

7. Check E. Nerve inflammation halts trial for

Alzheimer's drug. Nature 2002;415:462.

8. Cirrito JR, May PC, O'Dell MA, Taylor JW,

Parsadanian M, Cramer JW, Audia JE, Nissen JS,

Bales KR, Paul SM, DeMattos RB, Holtzman DM.

In vivo assessment of brain interstitial fluid with

microdialysis reveals plaque-associated changes in

amyloid-beta metabolism and half-life. J Neurosci

2003;23:8844-53.

9. Crouch PJ, Hung LW, Adlard PA, Cortes M, Lal V,

Filiz G, Perez KA, Nurjono M, Caragounis A, Du T,

Laughton K, Volitakis I, Bush AI, Li QX, Masters

CL, Cappai R, Cherny RA, Donnelly PS, White AR,

Barnham KJ. Increasing Cu bio-availability inhibits

Abeta oligomers and tau phosphorylation. Proc Natl

Acad Sci USA. 2009; 106:381-6.

10. Dawson GR, Seabrook GR, Zheng H, Smith DW,

Graham S, O'Dowd G, Bowery BJ, Boyce S,

Trumbauer ME, Chen HY, Van der Ploeg LH,

Sirinathsinghji DJ. Age-related cognitive deficits,

impaired long-term potentiation and reduction in

synaptic marker density in mice lacking the beta-

amyloid precursor protein. Neuroscience 1999;90: 1-

13.

11. De Bona P, ML Giuffrida , Caraci F, Copani A,

Pignataro B, Attanasio F, Cataldo S, Pappalardo G,

Rizzarelli E. Design and synthesis of new trehalose-

conjugated pentapeptides as inhibitors of Abeta(1-

42) fibrillogenesis and toxicity. J Pept Sci

2009;15:220-8.

12. de la Monte SM, Wands JR. Review of insulin and

insulin-like growth factor expression, signaling, and

malfunction in the central nervous system: relevance

to Alzheimer's disease. J Alzheimers Dis 2005;7:45-

61.

13. Dineley KT, Bell KA, Bui D, Sweatt JD. beta -

Amyloid peptide activates alpha 7 nicotinic

acetylcholine receptors expressed in Xenopus

oocytes. J Biol Chem 2002;277:25056-61.

14. Doble A. The role of excitotoxicity in neurode-

generative disease: implications for therapy.

Pharmacol Ther 1999;81:163-21.

15. Doig AJ. Peptide inhibitors of beta-amyloid

aggregation. Curr Opin Drug Discov Devel 2007;

10:533-9.

16. Eckman EA, Eckman CB. Abeta-degrading

enzymes: modulators of Alzheimer's disease path-

ogenesis and targets for therapeutic intervention.

Biochem Soc Trans 2005;33:1101-5.

17. Fleisher AS, Raman R, Siemers ER, Becerra L,

Clark CM, Dean RA, Farlow MR, Galvin JE,

Peskind ER, Quinn JF, Sherzai A, Sowell BB, Aisen

PS, Thal LJ. Phase 2 safety trial targeting amyloid

beta production with a gamma-secretase inhibitor in

Alzheimer disease. Arch Neurol 2008; 65:1031-8.

18. Franke TF, Kaplan DR, Cantley LC. PI3K:

downstream AKTion blocks apoptosis. Cell. 1997;

88:435-7.

19. Garcia-Osta A, Alberini CM. Amyloid beta mediates

memory formation. Learn Mem. 2009; 16:267-72.

20. Giuffrida ML, Caraci F, Pignataro B, Cataldo S, De

Bona P, Bruno V, Molinaro G, Pappalardo G,

Messina A, Palmigiano A, Garozzo D, Nicoletti F,

Rizzarelli E, Copani A. Beta-amyloid monomers are

neuroprotective. J Neurosci 2009;29:10582-7.

21. Gong Y, Chang L, Viola KL, Lacor PN, Lambert

MP, Finch CE, Krafft GA, Klein WL. Alzheimer's

disease-affected brain: presence of oligomeric A beta

ligands (ADDLs) suggests a molecular basis for

reversible memory loss. Proc Natl Acad Sci USA

2003;100:10417-22.

22. Green RC, Schneider LS, Amato DA, Beelen AP,

Wilcock G, Swabb EA, Zavitz KH. Effect of

tarenflurbil on cognitive decline and activities of

MONOMER STATE OF BETA-AMYLOID

VOLUME 21, NO. 1, 2010

91

daily living in patients with mild Alzheimer disease:

a randomized controlled trial. JAMA.

2009;302:2557-64.

23. Gu Z, Liu W, Yan Z. {beta}-Amyloid impairs

AMPA receptor trafficking and function by reducing

Ca2+/calmodulin-dependent protein kinase II

synaptic distribution. J Biol Chem 2009;284:10639-

49.

24. Hansson O, Zetterberg H, Buchhave P, Andreasson

U, Londos E, Minthon L, Blennow K. Prediction of

Alzheimer's disease using the CSF Abeta42/Abeta40

ratio in patients with mild cognitive impairment.

Dement Geriatr Cogn Disord 2007;23:316-20.

25. Hardy J. The shorter amyloid cascade hypothesis.

Neurobiol Aging.1999;20:85;discussion 87.

26. Harrison SM, Harper AJ, Hawkins J, Duddy G, Grau

E, Pugh PL, Winter PH, Shilliam CS, Hughes ZA,

Dawson LA, Gonzalez MI, Upton N, Pangalos MN,

Dingwall C. BACE1 (beta-secretase) transgenic and

knockout mice: identifi-cation of neurochemical

deficits and behavioral changes. Mol Cell Neurosci

2003;24:646-55.

27. Holmes C, Boche D, Wilkinson D, Yadegarfar G,

Hopkins V, Bayer A, Jones RW, Bullock R, Love S,

Neal JW, Zotova E, Nicoll JA. Long-term effects of

Abeta42 immunisation in Alzheimer's disease:

follow-up of a randomised, placebo-controlled phase

I trial. Lancet 2008;372:216-23.

28. Kamenetz F, Tomita T, Hsieh H, Seabrook G,

Borchelt D, Iwatsubo T, Sisodia S, Malinow R . APP

processing and synaptic function. Neuron

2003;37:925-37.

29. Kim HJ, Chae SC, Lee DK, Chromy B, Lee SC,

Park YC, Klein WL, Krafft GA, Hong ST. Selective

neuronal degeneration induced by soluble oligomeric

amyloid beta protein. FASEB J 2003;17:118-20.

30. Klyubin I, Walsh DM, Lemere CA, Cullen WK,

Shankar GM, Betts V, Spooner ET, Jiang L, Anwyl

R, Selkoe DJ, Rowan MJ. Amyloid beta protein

immunotherapy neutralizes Abeta oligomers that

disrupt synaptic plasticity in vivo. Nat Med

2005;11:556-61.

31. Kokjohn TA, Roher AE. Antibody responses,

amyloid-beta peptide remnants and clinical effects of

AN-1792 immunization in patients with AD in an

interrupted trial. CNS Neurol Disord Drug Targets

2009;8:88-97.

32. Lambert MP, Velasco PT, Viola KL, Klein WL.

Targeting generation of antibodies specific to

conformational epitopes of amyloid beta-derived

neurotoxins. CNS Neurol Disord Drug Targets

2009;8:65-81.

33. Lambert MP, Barlow AK, Chromy BA, Edwards C,

Freed R, Liosatos M, Morgan TE, Rozovsky I,

Trommer B, Viola KL, Wals P, Zhang C, Finch CE,

Krafft GA, Klein WL. Diffusible, nonfibrillar

ligands derived from Abeta1-42 are potent central

nervous system neurotoxins. Proc Natl Acad Sci

USA 1998;95:6448-53.

34. Lannfelt L, Blennow K, Zetterberg H, Batsman S,

Ames D, Harrison J, Masters CL, Targum S, Bush

AI, Murdoch R, Wilson J, Ritchie CW. Safety,

efficacy, and biomarker findings of PBT2 in

targeting Abeta as a modifying therapy for

Alzheimer's disease: a phase IIa, double-blind,

randomised, placebo-controlled trial. Lancet Neurol

2008;7:779-86.

35. Lee DH, Wang HY. Differential physiologic

responses of alpha7 nicotinic acetylcholine receptors

to beta-amyloid1-40 and beta-amyloid1-42. J

Neurobiol 2003;55:25-30.

36. Lemere CA. Developing novel immunogens for a

safe and effective Alzheimer's disease vaccine. Prog

Brain Res 2009;175:83-93.

37. Leow AD et al. Alzheimer's disease neuroimaging

initiative: a one-year follow up study using tensor-

based morphometry correlating degenerative rates,

biomarkers and cognition. Neuroimage. 2009;45:

645-55.

38. Lesne S, Koh MT, Kotilinek L, Kayed R, Glabe CG,

Yang A, Gallagher M, Ashe KH. A specific

amyloid-beta protein assembly in the brain impairs;

memory. Nature 2006;440:352-7.

39. Li L, Holscher C. Common pathological processes in

Alzheimer disease and type 2 diabetes: a review.

Brain Res Rev 2007;56:384-402.

40. Lue LF, Kuo YM, Roher AE, Brachova L, Shen Y,

Sue L, Beach T, Kurth JH, Rydel RE, Rogers J.

Soluble amyloid beta peptide concentration as a

predictor of synaptic change in Alzheimer's disease.

Am J Pathol 1999;155:853-62.

41. Mackic JB, Stins M, McComb JG, Calero M, Ghiso

J, Kim KS, Yan SD, Stern D, Schmidt AM,

Frangione B, Zlokovic BV. Human blood-brain

barrier receptors for Alzheimer's amyloid-beta 1- 40.

Asymmetrical binding, endocytosis, and trans-

cytosis at the apical side of brain microvascular

endothelial cell monolayer. J Clin Invest 1998;

102:734-743.

42. McLean CA, Cherny RA, Fraser FW, Fuller SJ,

Smith MJ, Beyreuther K, Bush AI, Masters CL.

Soluble pool of Abeta amyloid as a determinant of

severity of neurodegeneration in Alzheimer's

disease. Ann Neurol 1999;46:860-6.

43. Mollenhauer B, Bibl M, Trenkwalder C, Stiens G,

Cepek L, Steinacker P, Ciesielczyk B, Neubert K,

M.L. GIUFFRIDA ET AL.

REVIEWS IN THE NEUROSCIENCES

92

Wiltfang J, Kretzschmar HA, Poser S, Otto M.

Follow-up investigations in cerebrospinal fluid of

patients with dementia with Lewy bodies and

Alzheimer's disease. J Neural Transm 2005;112:

933-48.

44. Moloney AM, Griffin RJ, Timmons S, O'Connor R,

Ravid R, O'Neill C Defects in IGF-1 receptor,

insulin receptor and IRS-1/2 in Alzheimer's disease

indicate possible resistance to IGF-1 and insulin

signalling. Neurobiol Aging 2008;31:224-43.

45. Muller U, Cristina N, Li ZW, Wolfer DP, Lipp HP,

Rulicke T, Brandner S, Aguzzi A, Weissmann C.

Behavioral and anatomical deficits in mice

homozygous for a modified beta-amyloid pre-cursor

protein gene. Cell. 1994;79:755-65.

46. Nicoll JA, Wilkinson D, Holmes C, Steart P,

Markham H, Weller RO. Neuropathology of human

Alzheimer disease after immunization with amyloid-

beta peptide: a case report. Nat Med 2003;9:448-52.

47. Noguchi M, Yoshita M, Matsumoto Y, Ono K,

Iwasa K, Yamada M. Decreased beta-amyloid

peptide42 in cerebrospinal fluid of patients with

progressive supranuclear palsy and corticobasal

degeneration. J Neurol Sci 2005;237:61-5.

48. Oddo S, LaFerla FM. The role of nicotinic

acetylcholine receptors in Alzheimer's disease. J

Physiol Paris 2006;99:172-9.

49. Ohno M, Sametsky EA, Younkin LH, Oakley H,

Younkin SG, Citron M, Vassar R, Disterhoft JF.

BACE1 deficiency rescues memory deficits and

cholinergic dysfunction in a mouse model of

Alzheimer's disease. Neuron 2004;41:27-33.

50. Otto M, Esselmann H, Schulz-Shaeffer W, Neumann

M, Schroter A, Ratzka P, Cepek L, Zerr I, Steinacker

P, Windl O, Kornhuber J, Kretz-schmar HA, Poser

S, Wiltfang J. Decreased beta-amyloid1-42 in

cerebrospinal fluid of patients with Creutzfeldt-

Jakob disease. Neurology 2000; 54:1099-102.

51. Palop JJ, Mucke L. Epilepsy and cognitive

impairments in Alzheimer disease. Arch Neurol

2009;66:435-40.

52. Palop JJ, Chin J, Roberson ED, Wang J, Thwin MT,

Bien-Ly N, Yoo J, Ho KO, Yu GQ, Kreitzer A,

Finkbeiner S, Noebels JL, Mucke L. Aberrant

excitatory neuronal activity and compensatory

remodeling of inhibitory hippocampal circuits in

mouse models of Alzheimer's disease. Neuron

2007;55:697-711.

53. Panza F, Solfrizzi V, Frisardi V, Capurso C,

D'Introno A, Colacicco AM, Vendemiale G,

Capurso A, Imbimbo BP. Disease-modifying

approach to the treatment of Alzheimer's disease:

from alpha-secretase activators to gamma-secretase

inhibitors and modulators. Drugs Aging

2009;26:537-55.

54. Patton RL, Kalback WM, Esh CL, Kokjohn TA,

Van Vickle GD, Luehrs DC, Kuo YM, Lopez J,

Brune D, Ferrer I, Masliah E, Newel AJ, Beach TG,

Castano EM, Roher AE. Amyloid-beta peptide

remnants in AN-1792-immunized Alzheimer's

disease patients: a biochemical analysis. Am J Pathol

2006;169:1048-63.

55. Pettit DL, Shao Z, Yakel JL. beta-Amyloid(1-42)

peptide directly modulates nicotinic receptors in the

rat hippocampal slice. J Neurosci 2001;21: RC120.

56. Plant LD, Boyle JP, Smith IF, Peers C, Pearson HA.

The production of amyloid beta peptide is a critical

requirement for the viability of central neurons. J

Neurosci 2003;23:5531-5.

57. Puzzo D, Privitera L, Leznik E, Fa M, Staniszewski

A, Palmeri A, Arancio O. Picomolar amyloid-beta

positively modulates synaptic plasticity and memory

in hippocampus. J Neurosci 2008;28:14537-45.

58. Qiu WQ, Folstein MF. Insulin, insulin-degrading

enzyme and amyloid-beta peptide in Alzheimer's

disease: review and hypothesis. Neurobiol Aging

2006;27:190-8.

59. Roselli F, Tirard M, Lu J, Hutzler P, Lamberti P,

Livrea P, Morabito M, Almeida OF. Soluble beta-

amyloid1-40 induces NMDA-dependent degrada-

tion of postsynaptic density-95 at glutamatergic

synapses. J Neurosci 2005;25:11061-70.

60. Rowan MJ, Klyubin I, Wang Q, Anwyl R. Synaptic

plasticity disruption by amyloid beta protein:

modulation by potential Alzheimer's disease

modifying therapies. Biochem Soc Trans

2005;33:563-67.

61. Sabayan B, Foroughinia F, Mowla A, Borhani-

haghighi A. Role of insulin metabolism disturbances

in the development of Alzheimer disease: mini

review. Am J Alzheimers Dis Other Demen 2008;

23:192-9.

62. Salloway S, Sperling R, Gilman S, Fox NC,

Blennow K, Raskind M, Sabbagh M, Honig LS,

Doody R, van Dyck CH, Mulnard R, Barakos J,

Gregg KM, Liu E, Lieberburg I, Schenk D, Black R,

Grundman M. A phase 2 multiple ascending dose

trial of bapineuzumab in mild to moderate

Alzheimer disease. Neurology. 2009;73:2061-70.

63. Schenk D et al. Immunization with amyloid-beta

attenuates Alzheimer-disease-like pathology in the

PDAPP mouse. Nature 1999;400:173-77.

64. Senechal Y, Kelly PH, Dev KK. Amyloid precursor

protein knockout mice show age-dependent deficits

in passive avoidance learning. Behav Brain Res

2008;186:126-32.

MONOMER STATE OF BETA-AMYLOID

VOLUME 21, NO. 1, 2010

93

65. Sensi SL, Paoletti P, Bush AI, Sekler I. Zinc in the

physiology and pathology of the CNS. Nat Rev

Neurosci 2009;10:780-91.

66. Sjogren M, Davidsson P, Wallin A, Granerus AK,

Grundstrom E, Askmark H, Vanmechelen E,

Blennow K. Decreased CSF-beta-amyloid 42 in

Alzheimer's disease and amyotrophic lateral sclerosis

may reflect mismetabolism of beta-amyloid induced

by disparate mechanisms. Dement Geriatr Cogn

Disord 2002;13:112-8.

67. Snyder EM, Nong Y, Almeida CG, Paul S, Moran T,

Choi EY, Nairn AC, Salter MW, Lombroso PJ,

Gouras GK, Greengard P. Regulation of NMDA

receptor trafficking by amyloid-beta. Nat Neurosci

2005;8:1051-8.

68. Soto C, Kindy MS, Baumann M, Frangione B.

Inhibition of Alzheimer's amyloidosis by peptides

that prevent beta-sheet conformation. Biochem

Biophys Res Commun 1996;226:672-80.

69. Soucek T, Cumming R, Dargusch R, Maher P,

Schubert D. The regulation of glucose metabolism

by HIF-1 mediates a neuroprotective response to

amyloid beta peptide. Neuron 2003;39:43-56.

70. Sun X, Chiu CC, Liebson E, Crivello NA, Wang L,

Claunch J, Folstein M, Rosenberg I, Mwamburi DM,

Peter I, Qiu WQ. Depression and plasma amyloid

beta peptides in the elderly with and without the

apolipoprotein E4 allele. Alzheimer Dis Assoc

Disord 2009;23:238-44.

71. Teplow DB. Structural and kinetic features of

amyloid beta-protein fibrillogenesis. Amyloid

1998;5:121-42.

72. Town T. Alternative Abeta immunotherapy

approaches for Alzheimer's disease. CNS Neurol

Disord Drug Targets 2009;8:114-27.

73. Townsend M, Shankar GM, Mehta T, Walsh DM,

Selkoe DJ. Effects of secreted oligomers of amyloid

beta-protein on hippocampal synaptic plasticity: a

potent role for trimers. J Physiol 2006;572:477-92.

74. Vassar R, Kovacs DM, Yan R, Wong PC. The beta-

secretase enzyme BACE in health and Alzheimer's

disease: regulation, cell biology, function, and

therapeutic potential. J Neurosci 2009;29:12787-94.

75. Walsh DM, Klyubin I, Fadeeva JV, Cullen WK,

Anwyl R, Wolfe MS, Rowan MJ, Selkoe DJ.

Naturally secreted oligomers of amyloid beta protein

potently inhibit hippocampal long-term potentiation

in vivo. Nature 2002;416:535-9.

76. Wang H, Song L, Laird F, Wong PC, Lee HK.

BACE1 knock-outs display deficits in activity-

dependent potentiation of synaptic transmission at

mossy fiber to CA3 synapses in the hippocampus. J

Neurosci 2008;28:8677-81.

77. Wang HW, Pasternak JF, Kuo H, Ristic H, Lambert

MP, Chromy B, Viola KL, Klein WL, Stine WB,

Krafft GA, Trommer BL. Soluble oligomers of beta

amyloid (1-42) inhibit long-term potentiation but not

long-term depression in rat dentate gyrus. Brain Res

2002;924:133-40.

78. West AE, Griffith EC, Greenberg ME. Regulation of

transcription factors by neuronal activity. Nat Rev

Neurosci 2002;3:921-31.

79. Whitson JS, Selkoe DJ, Cotman CW. Amyloid beta

protein enhances the survival of hippocampal

neurons in vitro. Science 1989;243:1488-90.

80. Wilcock GK, Black SE, Hendrix SB, Zavitz KH,

Swabb EA, Laughlin MA. Efficacy and safety of

tarenflurbil in mild to moderate Alzheimer's disease:

a randomised phase II trial. Lancet Neurol

2008;7:483-93.

81. Wisniewski T, Konietzko U. Amyloid-beta

immunisation for Alzheimer's disease. Lancet Neurol

2008;7:805-11.

82. Xia W, Yang T, Shankar G, Smith IM, Shen Y,

Walsh DM, Selkoe DJ. A specific enzyme-linked

immunosorbent assay for measuring beta-amyloid

protein oligomers in human plasma and brain tissue

of patients with Alzheimer disease. Arch Neurol

2009;66:190-9.

83. Xia W, Zhang J, Kholodenko D, Citron M, Podlisny

MB, Teplow DB, Haass C, Seubert P, Koo EH,

Selkoe DJ. Enhanced production and

oligomerization of the 42-residue amyloid beta-

protein by Chinese hamster ovary cells stably

expressing mutant presenilins. J Biol Chem 1997;

272:7977-82.

84. Zhao WQ, Lacor PN, Chen H, Lambert MP, Quon

MJ, Krafft GA, Klein WL. Insulin receptor

dysfunction impairs cellular clearance of neurotoxic

oligomeric a{beta}. J Biol Chem 2009; 284:18742-

53.

85. Zhao WQ, De Felice FG, Fernandez S, Chen H,

Lambert MP, Quon MJ, Krafft GA, Klein WL.

Amyloid beta oligomers induce impairment of

neuronal insulin receptors. FASEB J 2008;22: 246-

60.