Skip to main content
Journal of Biomedicine and Biotechnology logoLink to Journal of Biomedicine and Biotechnology
. 2011 Aug 23;2011:697036. doi: 10.1155/2011/697036

A Window into the Heterogeneity of Human Cerebrospinal Fluid Aβ Peptides

Roberta Ghidoni 1,*, Anna Paterlini 1, Valentina Albertini 1, Elena Stoppani 1, Giuliano Binetti 2, Kjell Fuxe 3, Luisa Benussi 2, Luigi F Agnati 4
PMCID: PMC3163146  PMID: 21876644

Abstract

The initiating event in Alzheimer's disease (AD) is an imbalance in the production and clearance of amyloid beta (Aβ) peptides leading to the formation of neurotoxic brain Aβ assemblies. Cerebrospinal Fluid (CSF), which is a continuum of the brain, is an obvious source of markers reflecting central neuropathologic features of brain diseases. In this review, we provide an overview and update on our current understanding of the pathobiology of human CSF Aβ peptides. Specifically, we focused our attention on the heterogeneity of the CSF Aβ world discussing (1) basic research studies and what has been translated to clinical practice, (2) monomers and other soluble circulating Aβ assemblies, and (3) communication modes for Aβ peptides and their microenvironment targets. Finally, we suggest that Aβ peptides as well as other key signals in the central nervous system (CNS), mainly involved in learning and hence plasticity, may have a double-edged sword action on neuron survival and function.

1. Introduction

The “amyloid cascade hypothesis” suggests that the initiating event in Alzheimer's disease (AD) is an imbalance in the production and clearance of amyloid beta (Aβ) peptides leading to the formation of neurotoxic soluble and insoluble brain Aβ assemblies [1, 2]. Thus, Aβ has become a major therapeutic target, with various anti-Aβ strategies being pursued [3]. Biologically, monomeric Aβ is formed through the enzymatic cleavage of the transmembrane amyloid precursor protein (APP). The discovery of the APP gene was followed by the identification of missense mutations associated with familial, early-onset AD. These mutations are found in and around the Aβ region of APP (http://www.molgen.ua.ac.be/ADmutations/) and affect the production or aggregation properties of Aβ. The physiopathological processing of APP involves various proteolytic activities leading to a complex set of Aβ fragments. Full-length Aβ1-40 and Aβ1-42 peptides are generated by sequential proteolytic processing involving β and γ-secretases on APP [4]. These peptides (i.e., Aβ1-40, Aβ1-42) have been the dominant focus of research, but it is well established that N- and C-terminally truncated or modified forms of Aβ peptides also exist in AD brains [59]. The detection of N-terminal truncated Aβ peptides (especially Aβx-42) in young Down's syndrome and in preclinical AD brains suggests that the amino-truncated species are implicated in the very first step of amyloidosis [1012]. These forms are generated mainly by cleavage of APP between residues 16 and 17 of the Aβ domain via the α-secretase and by the alternative β′ cleavage of APP triggered by the β-secretase β-site APP-cleaving enzyme (BACE)1 [1315]. Heterogeneity at the C-terminus of Aβ also contributes to the molecular variety of Aβ peptides; according to some reports, due to its imprecise cleavage specificity, γ-secretase generates Aβ peptides of variable length at the C-terminus [16]. Recently, γ-secretase has also been shown to cleave near the cytoplasmic membrane boundary of APP, called ε-site cleavage [17]. In addition, it has been recently demonstrated that the combined activity of α- and β-secretases may generate the shortest forms (i.e., Aβ 1-15, Aβ 1-16) of C-terminally truncated Aβ peptides [18]. Body fluids, such as cerebrospinal fluid (CSF), plasma, serum, or urine represent a cellular protein-rich information reservoir that contains traces of what has been secreted into these fluids. In particular, CSF, which is a continuum of the brain, is an obvious source of markers reflecting central neuropathologic features of the brain diseases.

This review provides an overview and update on our current understanding of the pathobiology of human CSF Aβ peptides.

2. CSF Aβ Peptides in Translational Research

Has knowledge on pathobiology of Aβ been somehow translated to clinical practice? The criteria for the clinical diagnosis of AD were established by the National Institute of Neurological and Communicative Disorders and Stroke (NINCDS) and the Alzheimer's Disease and Related Disorders Association (ADRDA) workgroup in 1984 [19]. However, in the intervening 27 years, important advances in our understanding of AD, in our ability to detect the pathophysiological process of AD, and changes in conceptualization regarding the clinical spectrum of the disease have occurred [20, 21].

The revised diagnostic criteria proposed in 2011 by the National Institute of Aging and the Alzheimer's Association workgroup include the incorporation of biomarkers of the underlying disease state and formalization of different stages of disease—“preclinical AD,” “mild cognitive impairment (MCI) due to AD,” and “AD dementia”—in the diagnostic criteria [2224]. Biomarkers are parameters (physiological, biochemical, anatomic) that can be measured in vivo and that reflect specific features of disease-related pathophysiological processes. In recent years, a number of reports have utilised specific protein/peptide quantitation techniques such as ELISA to study the levels of selective moieties in CSF as biomarkers of this neurodegenerative disorder. The three major alterations in AD brain are extracellular amyloid plaques, axonal degeneration, and intraneuronal tangles, which can be monitored with the CSF biomarkers Aβ1-42, total tau, and phosphorylated tau, respectively. The onset and progression of AD biomarkers likely follows an ordered temporal pattern. Biomarkers of Aβ amyloid are indicative of initiating or upstream events which seem to be most dynamic (i.e., deviate most significantly from normal) before clinical symptoms. Biomarkers of neuronal injury and neuronal dysfunction are indicative of downstream pathophysiological processes which become dynamic later. There is evidence suggesting that combined assessment of CSF tau and Aβ1-42 have high diagnostic accuracy for established AD [25]. They may also be used to identify AD before onset of dementia at the stage of MCI, as shown in both mono-center and large-scale heterogeneous multicenter studies [2630]. Since CSF levels of the shorter Aβ1-40 isoform are unchanged or increased in AD, it has been proposed that measurement of the Aβ1-42/Aβ1-40 ratio might be superior to Aβ1-42 alone [3134]. Of note, Aβ1-42 is associated with impairment of cognitive function from a potentially early to a later disease phase [3537]. Decreased CSF Aβ1-42 is also seen in other neurodegenerative disorders [38]. Recent studies have shown associations between shorter forms of Aβ peptides and specific dementias: decreased Aβ1-38 levels correlated with frontotemporal dementia [39] and Aβ1-37 with Lewy Body dementia [40]. Thus, the detection of the whole spectrum of Aβ peptides in the CSF could be useful in order to improve early differential diagnosis.

3. The Large Family of CSF Aβ Peptides: The Mass Spectrometry-Based Detection

The predominant protein component of amyloid plaques are strongly aggregating peptides with an approximate molecular mass of 4 kDa. The main plaques component is the 42 amino acid isoform of Aβ; this isoform is highly hydrophobic and forms oligomers and fibrils that accumulate in extracellular plaques [41]. The deposition of the peptide in plaques is considered the underlying basis for the decrease in CSF Aβ1-42 levels seen in AD and incorporated in the new diagnostic criteria. In addition, other isoforms of Aβ, for example, pyro Aβ3-42, Aβ4-42, pyro Aβ11-42, Aβ17-42, Aβ1-40, and Aβ11-40 have been detected in the brains of sporadic AD and familial AD cases [512, 4246]. Aβ peptides heterogeneity is observed also in the human CSF (see Table 1) [4758]. The proteolytically processed Aβ peptides, however, are difficult to detect in the CSF-using standard methods, possibly because they comprise a heterogeneous set of both N- and C-terminally truncated peptides, some of which are present only at low levels. Many investigators used mass spectrometry (MS) for studying human CSF Aβ peptides. MS allows for the detection of a variety of modified and truncated Aβ peptides, thus enabling a more detailed and unbiased analysis of fragments that may play a role in neurodegeneration. The two main approaches are (1) the use of preactivated chip arrays that allow coupling with specific antibodies combined with surface-enhanced laser desorption and ionization time-of-flight (SELDI-TOF) MS (2) immunoprecipitation combined with matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF) MS. An immunoproteomic approach—which combines specificity of 6E10 (against Aβ epitope 1-16) mAb capture with precision of spectral analysis (i.e., SELDI-TOF MS)—has recently been successfully used to analyze Aβ peptides in human CSF; Maddalena et al. [50] detected 9 C-terminally and 1 N-terminally truncated Aβ peptides in CSF of AD patients and healthy controls subjects. while, with an analogous protocol, 10 Aβ fragments were found by Lewczuk et al. [55, 58]. Immunoprecipitation experiments employing 4G8 mAb and MALDI-MS analyses of Aβ peptides from 1 mL CSF revealed the presence of two previously unidentified N-terminally truncated Aβ peptides (i.e., Aβ11-30, Aβ11-40), along with a number of C-terminally truncated forms [47, 48]. Since 6E10 and 4G8 mAbs bind different portions of Aβ sequence, we tested whether the combined use of these two mAbs could improve the capture of N and C-terminally truncated Aβ peptides; of note, applying this optimized immunoproteomic assay—that employs very low sample volume (5 μL of CSF for each spot)—we detected a total of 15 Aβ peptides (12 C-terminally and 3 N-terminally truncated forms) in human CSF [51].

Table 1.

Summary of Aβ peptides in human CSF.

Aβ Peptides Theoretical mass* (Da) Literature
Aβ1-12 1424.61 [47]
Aβ1-13 1561.67 [4749]
Aβ2-14 1583.70 [50]
Aβ1-14 1698.73 [4749]
Aβ1-15 1826.78 [4749]
Aβ3-17 1881.90 [48]
Aβ2-17 1952.94 [48]
Aβ1-16 1954.88 [4749]
Aβ1-17 2067.96 [4752]
Aβ1-18 2167.03 [4751]
Aβ11-30 2212.11 [47]
Aβ1-19 2314.10 [4749, 51, 52]
Aβ1-20 2461.17 [4749]
Aβ6-27 2521.16 [53]
Aβ11-34 2608.39 [53]
Aβ1-27 3133.44 [53]
Aβ11-40 3150.68 [47, 51, 54]
Aβ6-34 3167.60 [53]
Aβ1-28 3261.53 [47, 48, 53, 55]
Aβ6-35 3298.63 [53]
Aβ12-43 3306.80 [53]
Aβ10-40 3313.74 [51, 54]
Aβ1-29 3318.56 [55]
Aβ11-42 3334.80 [51, 54, 56]
Aβ1-30 3389.59 [4749, 53]
Aβ11-43 3435.85 [53]
Aβ3-34 3599.80 [53]
Aβ1-33 3672.78 [47, 4951, 54, 55, 57]
Aβ1-34 3785.87 [47, 4951, 5355, 57]
Aβ1-35 3916.91 [51, 53, 57]
Aβ1-36 4015.98 [51]
Aβ1-37 4073.00 [47, 4951, 54, 55, 57, 58]
Aβ1-38 4130.02 [47, 4951, 5355, 57, 58]
Aβ1-39 4229.09 [47, 4951, 54, 55, 57, 58]
Aβ1-40 4328.16 [47, 4951, 5355, 57, 58]
Aβ1-42 4512.28 [47, 4951, 5458]
Aβ3-44 4526.33 [58]
Aβ1-45 or Aβ2-46 4825.48 or 4809.52 [55]
Aβ3-47 4851.56 [58]

*The masses presented are the monoisotopic protonated molecules.

In addition, we determined mass profiles of Aβ peptides in the CSF of patients carrying familial AD-associated mutations (i.e., APP T719P, PS1 P117L, and PS2 T122R); these mutations were associated with an overall reduction of Aβ species. Interestingly, the APP T719P mutation unbalanced the relative proportion of Aβ peptides with a reduction of Aβ1-40 and Aβ1-42 paralleled by an increase of Aβ1-38 and Aβ10-40 [54]. In accordance with these data, Portelius and coauthors [49] reported a reduction C-terminally truncated Aβ peptides in CSF of affected and unaffected subjects carrying PS1 A431E mutation. An unbalance of Aβ isoforms was also detected in CSF of sporadic AD and MCI patients [50, 52, 56, 57]. Interestingly, within a phase II clinical trial, it has been recently demonstrated that Aβ1-14, Aβ1-15, and Aβ1-16 are positive and very sensitive biomarkers for γ-secretase inhibition (even at doses that do not affect Aβ1-42 or Aβ1-40) [59]. Thus, Aβ isoforms may be novel biomarkers to monitor the onset and progression of cognitive decline and the biochemical effect of disease-modifying drugs in AD clinical trials.

4. Beyond Aβ Monomers: CSF Circulating Aβ Oligomers

In the human brain it is likely that multiple Aβ assemblies, that are in dynamic equilibrium almost simultaneously, alter brain cell function and that different toxic effects may occur virtually concurrently in various regions of the cerebrum. Several lines of evidence have converged to demonstrate that soluble oligomers of Aβ may be responsible for synaptic dysfunction in AD animal models and in the brains of AD patients [46, 60, 61]. Small diffusible Aβ oligomers have been shown to exert neurotoxic effects in cultured neurons [6264]. It has been hypothesized that such prefibrillar assemblies might also be neurotoxic in vivo since synaptic, electrophysiological, and behavioral changes have been well documented in young APP transgenic mice before plaque formation [65, 66]. Accordingly, soluble Aβ oligomers have been found to block, in vivo, hippocampal long-term potentiation (LTP), a synaptic correlate of memory and learning [6771]. Importantly, Aβ immunotherapy can protect against the neuropathology and cognitive deficits observed in APP transgenic mice and also prevent the LTP inhibition induced by Aβ oligomers [68]. Soluble oligomeric Aβ has been shown to be present in human CSF [7274]. Human derived soluble Aβ seems to have a pathophysiological role in the brain; the CSF-derived Aβ dimers—and not the monomers—potently disrupt synaptic plasticity in vivo [75]. Of note, it has been reported that CSF circulating oligomers are increased in AD and MCI patients, and their levels are negatively correlated with Mini-Mental State Examination scores [76, 77]. Thus, an emerging strategy within the AD field is to use oligomeric Aβ as a possible biomarker/therapeutic target for the disease. The actual identity of the oligomer participating in AD pathogenesis remains elusive although several lines of evidence suggest that AD-associated oligomers are primarily composed of Aβ42. Nevertheless Gao and coworkers, using a novel misfolded protein assay, found an enrichment of Aβ40-containing oligomers in AD CSF [78] and suggested these assemblies as biomarker for early diagnosis of AD. Although Aβ oligomers are attractive AD biomarker candidates, several issues relating to these molecules persist. The levels of these Aβ species in CSF seem to be very low in comparison with Aβ monomers and the precise molecular identity of these soluble toxins remains unsettled; thus more precise mass spectrometry analyses are needed in order to better characterize the molecular weight and composition of the most neurotoxic species. Furthermore, assays suitable for large clinical studies are still to be developed for these molecules. The development of conformation-sensitive antibody domains targeting the Aβ oligomers [7983] is of great interest for research in this field. Targeting the pathological assemblies of Aβ with specific probes, for mechanistic studies, for intracellular imaging, or for therapeutic purposes, is therefore very important.

5. Aβ Peptides Are Double-Edged Sword Signals Transmitted Both via Volume and Wiring Transmission

As discussed above, Aβ peptides have been regarded as the principal toxic factor in the neurodegeneration of AD. Intense research effort has, therefore, been directed at determining their sources, activities, and fates, primarily with a view of preventing their formation or toxic actions, or promoting their degradation.

These are important studies and very promising ones for a better understanding of the pathogenesis of AD. However, in our opinion, a crucial aspect is the discovery of the physiological role of these peptides.

Thus, the following points will be briefly discussed as far as the Aβ peptides are concerned:

  1. communication modes for these peptides, hence (volume transmission (VT)) versus (wiring transmission (WT)) versus (VT and WT);

  2. micro-environment where the targets for Aβ peptides are located, hence plasma membrane versus intracellular environment;

  3. possible physiological roles of Aβ peptides.

Finally, a previously published theoretical proposal [84] will be summarised since it can give a possible frame for interpreting otherwise contradictory data on Aβ peptides functions. The hypothesis is based on the concept that Aβ peptides as well as other key signals in the central nervous system (CNS) mainly involved in learning, and hence plasticity may have a double-edged sword action on neuron survival and function.

5.1. Communication Modes for Aβ Peptides and Their Microenvironment Targets

It has been proposed that two main modes for intercellular communication are in operation in the CNS, namely, the VT and the WT [85].

The characteristics of the channel connecting two nodes of the network, that is, the cell source of the signal with the cell-target of the signal allow distinguishing the VT from the WT.

(i) VT is characterized by a channel with a poorly defined physical substrate and signal transmission takes place via diffusion (or vector migration) in the medium interposed between nodes. Recently, it has been shown that several messages can be sent via microvesicles (acting as protective containers hence like the bag of a roamer), dispatched into the extracellular space (ECS) and diffusing until the proper targets are reached [8688].

Different types of microvesicles have been described, which are the result of specific cellular phenomena [86]. In particular, exosomes are microvesicles contained within a special class of membrane-bound organelles (endosomes), which can be released by fusion of the limiting membrane of the MVB with the plasma membrane.

(ii) WT is characterized by the transmission of the signal along a channel with a well-defined physical substrate; thus, a “wire” links the source node with the target node. Classically, in the case of neural networks, the WT-channel is formed by an axon and a chemical synapse.

However, two more subclasses of WT play a role in the CNS. The first one is represented by the well-characterized gap junctions, while the second one, the clear-cut in vivo demonstration of which has not yet been provided, is represented by the tunnelling nanotubes (TNTs) that are transient structures forming a “private” direct channel connecting two cells. They have a diameter of 50–200 nm and a length up to several cell diameters. Several in vitro studies demonstrated that these structures make possible the exchange of proteins, mtDNA, RNA, and whole organelles between cells [89]. It is interesting to note that Aβ peptides can be transmitted according to both VT and WT. Actually, it has been shown that these signals can use several possible modes of intercellular communication:

  1. the classical VT mode that is diffusion in the ECS [9094],

  2. the Roamer Type of VT that is diffusion via exosomes [9599],

  3. the TNT mode of WT [100].

The targets for the Aβ peptides are located both at the plasma membrane level [101, 102] and at intracellular level where they may exert an “intracrine function” [95, 103, 104].

5.2. Possible Functional Roles of Aβ Peptides

We completely agree with Pearson and Peers' view that Aβ peptides should have important physiological roles and may even be crucial for neuronal cell survival and CNS function. Thus, the view of Aβ being a purely toxic peptide requires a reevaluation [105]. In support of such a proposal, there are several papers, two of these will be cited since while the first one shows a role of Aβ peptides on learning [106], the other one opens a new field by giving evidence for a possible role of these peptides as antimicrobial agents [107].

Thus, it has been shown that, in contrast with its pathological role when accumulated, endogenous Aβ in normal hippocampi mediates learning and memory formation probably via nicotinic acetylcholine receptors. Furthermore, hippocampal injection of picomolar concentrations of exogenous Aβ1-42 enhances memory consolidation. Hence, Aβ peptides, including Aβ1-42, play an important physiological role in hippocampal memory formation.

As mentioned above, recently a new possible function for Aβ peptides has been demonstrated, namely, the antimicrobial action. Thus, it has been shown that many of the physiochemical and biological properties previously reported for Aβ are similar to those of a group of biomolecules collectively known as ‘‘antimicrobial peptides” (AMPs; also called ‘‘host defense peptides”) which function in the innate immune system. These peptides are potent, broad-spectrum antibiotics that target several infective agents. In particular, the pleiotropic LL-37 peptide is a widely expressed archetypal AMP present also in humans that exhibits striking similarities to Aβ, including a propensity to form cytotoxic soluble oligomers and insoluble fibrils with classical histochemical properties of tinctorial amyloid. Soscia et al. [107] findings reveal that Aβ exerts antimicrobial activity against eight common and clinically relevant microorganisms with a potency equivalent to, and in some cases greater than, LL-37. These findings obviously impose a great caution in developing future AD treatment strategies based on the drastic reduction of synthesis and levels of Aβ peptides.

5.3. Double-Edged Sword Action of Aβ Peptides on Neuron Plasticity and Survival

More than one century ago, Tanzi proposed that learning processes in the CNS are basically due to plastic changes of neuronal networks [108].

As pointed out by Taylor and Gaze, neuronal plasticity allowing continuous CNS adaptation to the challenges of the environment plays a fundamental role not only for learning processes. Actually, plasticity in the nervous system means a patterned or ordered alteration in structure and function brought about by development, experience, or injury [109].

Thus, this definition mentions age, learning, and lesions as factors triggering out plasticity.

In this paper the concept is introduced that physiological processes (such as learning and memory) as well as repairable processes (such as those occurring after lesions or during ageing), being all rooted in CNS rearrangements, are competing for the brain plasticity [110], which exists as a fixed amount (“total brain plasticity capability,” see [84]).

It has been demonstrated that some signals, such as excito-amino acids, Aβ peptides, and α-synuclein (α-syn), are not only involved in information handling by the neuronal circuits, but also trigger out CNS plasticity [84]. It has also been shown that these signals are potentially dangerous possibly since, interalia, they force the neuronal circuits to move from one stable state towards a new state. Several mechanisms are put in action to protect neurons and glial cells from these potentially harmful signals and hence favouring the emergence of only their physiological functions. However, ageing and neurodegenerative diseases, on one side, increase the need of plasticity for the CNS repair but, on the other side, cause a reduction in the secretion of several trophic factors (e.g., BDNF and NGF) leading to a less effective neuroprotection and deficits in neural plasticity [111, 112].

Against this background, it has been shown that in ageing and neurodegenerative diseases functionally ambivalent (i.e., double-edged sword) signals such as Aβ and α-syn are secreted at a high rate possibly in the attempt of maximizing neuronal plasticity. It has been proposed that in the long run these peptides do not exert their possible physiological actions but on the contrary may favour neurodegenerative processes.

Soscia et al. [107] have demonstrated that an increased Aβ generation/accumulation leading to AD pathology may be mediated by a response of the innate immune system to a perceived infection. This model is in agreement with data supporting a central role for neuroinflammation in AD neuropathology [113].

Thus, not only genetic factors may contribute to activation of the innate immune system by regulating Aβ production and clearance but also a transient infection may lead to a self-perpetuating innate immune response.

These findings allow an update of the hypothesis made in the JNT 2009 [84] (see Figure 1).

Figure 1.

Figure 1

Schematic representation of the ‘‘Push and Pull Control” of structural and functional plasticity of neuronal circuits and how this control is related to learning processes (plastic changes of the circuits) and maintenance of the memory traces (stability of the circuits). The possible actions of Aβ peptides as double-edged sword signals are indicated. Broken arrows indicate reduction or inhibition.

Acknowledgments

This work was supported by Grants from Ricerca Corrente, Italian Ministry of Health; AFaR; Fondazione CARIPLO 2009-2633.

References

  • 1.Masters CL, Simms G, Weinman NA, Multhaup G, McDonald BL, Beyreuther K. Amyloid plaque core protein in Alzheimer disease and Down syndrome. Proceedings of the National Academy of Sciences of the United States of America. 1985;82(12):4245–4249. doi: 10.1073/pnas.82.12.4245. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Hardy J, Selkoe DJ. The amyloid hypothesis of Alzheimer’s disease: progress and problems on the road to therapeutics. Science. 2002;297(5580):353–356. doi: 10.1126/science.1072994. [DOI] [PubMed] [Google Scholar]
  • 3.Lemere CA, Masliah E. Can Alzheimer disease be prevented by amyloid-β immunotherapy? Nature Reviews Neurology. 2010;6(2):108–119. doi: 10.1038/nrneurol.2009.219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Haass C, Schlossmacher MG, Hung AY, et al. Amyloid β-peptide is produced by cultured cells during normal metabolism. Nature. 1992;359(6393):322–325. doi: 10.1038/359322a0. [DOI] [PubMed] [Google Scholar]
  • 5.Saido TC, Iwatsubo T, Mann DMA, Shimada H, Ihara Y, Kawashima S. Dominant and differential deposition of distinct β-amyloid peptide species, AβN3(pE), in senile plaques. Neuron. 1995;14(2):457–466. doi: 10.1016/0896-6273(95)90301-1. [DOI] [PubMed] [Google Scholar]
  • 6.Iwatsubo T, Saido TC, Mann DMA, Lee VMY, Trojanowski JQ. Full-length amyloid-β(1-42(43)) and amino-terminally modified and truncated amyloid-β42(43) deposit in diffuse plaques. American Journal of Pathology. 1996;149(6):1823–1830. [PMC free article] [PubMed] [Google Scholar]
  • 7.Russo C, Saido TC, DeBusk LM, Tabaton M, Gambetti P, Teller JK. Heterogeneity of water-soluble amyloid β-peptide in Alzheimer’s disease and Down’s syndrome brains. FEBS Letters. 1997;409(3):411–416. doi: 10.1016/s0014-5793(97)00564-4. [DOI] [PubMed] [Google Scholar]
  • 8.Russo C, Schettini G, Saido TC, et al. Presenilin-1 mutations in Alzheimer’s disease. Nature. 2000;405(6786):531–532. doi: 10.1038/35014735. [DOI] [PubMed] [Google Scholar]
  • 9.Portelius E, Bogdanovic N, Gustavsson MK, et al. Mass spectrometric characterization of brain amyloid beta isoform signatures in familial and sporadic Alzheimer’s disease. Acta Neuropathologica. 2010;120(2):185–193. doi: 10.1007/s00401-010-0690-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Russo C, Salis S, Dolcini V, et al. Amino-terminal modification and tyrosine phosphorylation of carboxy-terminal fragments of the amyloid precursor protein in Alzheimer's disease and Down's syndrome brain. Neurobiology of Disease. 2001;8(1):173–180. doi: 10.1006/nbdi.2000.0357. [DOI] [PubMed] [Google Scholar]
  • 11.Sergeant N, Bombois S, Ghestem A, et al. Truncated beta-amyloid peptide species in pre-clinical Alzheimer’s disease as new targets for the vaccination approach. Journal of Neurochemistry. 2003;85(6):1581–1591. doi: 10.1046/j.1471-4159.2003.01818.x. [DOI] [PubMed] [Google Scholar]
  • 12.Liu K, Solano I, Mann D, et al. Characterization of Abeta11-40/42 peptide deposition in Alzheimer’s disease and young Down’s syndrome brains: implication of N-terminally truncated Abeta species in the pathogenesis of Alzheimer’s disease. Acta neuropathologica. 2006;112(2):163–174. doi: 10.1007/s00401-006-0077-5. [DOI] [PubMed] [Google Scholar]
  • 13.Sisodia SS. β-Amyloid precursor protein cleavage by a membrane-bound protease. Proceedings of the National Academy of Sciences of the United States of America. 1992;89(13):6075–6079. doi: 10.1073/pnas.89.13.6075. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Vassar R, Bennett BD, Babu-Khan S, et al. β-Secretase cleavage of Alzheimer’s amyloid precursor protein by the transmembrane aspartic protease BACE. Science. 1999;286(5440):735–741. doi: 10.1126/science.286.5440.735. [DOI] [PubMed] [Google Scholar]
  • 15.Liu K, Doms RW, Lee VMY. Glu11 site cleavage and N-terminally truncated Aβ production upon BACE overexpression. Biochemistry. 2002;41(9):3128–3136. doi: 10.1021/bi015800g. [DOI] [PubMed] [Google Scholar]
  • 16.Portelius E, Price E, Brinkmalm G, et al. A novel pathway for amyloid precursor protein processing. Neurobiology of Aging. 2009;32(6):1090–1098. doi: 10.1016/j.neurobiolaging.2009.06.002. [DOI] [PubMed] [Google Scholar]
  • 17.Kametani F. ε-secretase: reduction of amyloid precursor protein ε-site cleavage in Alzheimer’s disease. Current Alzheimer Research. 2008;5(2):165–171. doi: 10.2174/156720508783954776. [DOI] [PubMed] [Google Scholar]
  • 18.Portelius E, Zhang B, Gustavsson MK, et al. Effects of γ-secretase inhibition on the amyloid β isoform pattern in a mouse model of Alzheimer’s disease. Neurodegenerative Diseases. 2009;6(5-6):258–262. doi: 10.1159/000264639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.McKhann G, Drachman D, Folstein M. Clinical diagnosis of Alzheimer's disease: report of the NINCDS-ADRDA work group under the auspices of Department of Health and Human Services Task Force on Alzheimer's disease. Neurology. 1984;34(7):939–944. doi: 10.1212/wnl.34.7.939. [DOI] [PubMed] [Google Scholar]
  • 20.Dubois B, Feldman HH, Jacova C, et al. Research criteria for the diagnosis of Alzheimer’s disease: revising the NINCDS-ADRDA criteria. Lancet Neurology. 2007;6(8):734–746. doi: 10.1016/S1474-4422(07)70178-3. [DOI] [PubMed] [Google Scholar]
  • 21.Dubois B, Feldman HH, Jacova C, et al. Revising the definition of Alzheimer’s disease: a new lexicon. The Lancet Neurology. 2010;9(11):1118–1127. doi: 10.1016/S1474-4422(10)70223-4. [DOI] [PubMed] [Google Scholar]
  • 22.McKhann GM, Knopman DS, Chertkow H, et al. The diagnosis of dementia due to Alzheimer's disease: recommendations from the National Institute on Aging-Alzheimer's Association workgroups on diagnostic guidelines for Alzheimer's disease. Alzheimer's and Dementia. 2011;7(3):263–269. doi: 10.1016/j.jalz.2011.03.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Albert MS, DeKosky ST, Dickson D, et al. The diagnosis of mild cognitive impairment due to Alzheimer's disease: recommendations from the National Institute on Aging-Alzheimer's Association workgroups on diagnostic guidelines for Alzheimer's disease. Alzheimer's and Dementia. 2011;7(3):270–279. doi: 10.1016/j.jalz.2011.03.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Sperling RA, Aisen PS, Beckett LA, et al. Toward defining the preclinical stages of Alzheimer's disease: recommendations from the National Institute on Aging-Alzheimer's Association workgroups on diagnostic guidelines for Alzheimer's disease. Alzheimer's and Dementia. 2011;7(3):280–292. doi: 10.1016/j.jalz.2011.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Blennow K, Hampel H, Weiner M, Zetterberg H. Cerebrospinal fluid and plasma biomarkers in Alzheimer disease. Nature Reviews Neurology. 2010;6(3):131–144. doi: 10.1038/nrneurol.2010.4. [DOI] [PubMed] [Google Scholar]
  • 26.Mattsson N, Zetterberg H, Hansson O, et al. CSF biomarkers and incipient Alzheimer disease in patients with mild cognitive impairment. Journal of the American Medical Association. 2009;302(4):385–393. doi: 10.1001/jama.2009.1064. [DOI] [PubMed] [Google Scholar]
  • 27.Visser PJ, Verhey F, Knol DL, et al. Prevalence and prognostic value of CSF markers of Alzheimer’s disease pathology in patients with subjective cognitive impairment or mild cognitive impairment in the DESCRIPA study: a prospective cohort study. The Lancet Neurology. 2009;8(7):619–627. doi: 10.1016/S1474-4422(09)70139-5. [DOI] [PubMed] [Google Scholar]
  • 28.Shaw LM, Vanderstichele H, Knapik-Czajka M, et al. Cerebrospinal fluid biomarker signature in alzheimer’s disease neuroimaging initiative subjects. Annals of Neurology. 2009;65(4):403–413. doi: 10.1002/ana.21610. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Frisoni GB, Prestia A, Zanetti O, et al. Markers of Alzheimer’s disease in a population attending a memory clinic. Alzheimer’s and Dementia. 2009;5(4):307–317. doi: 10.1016/j.jalz.2009.04.1235. [DOI] [PubMed] [Google Scholar]
  • 30.Galluzzi S, Geroldi C, Ghidoni R, et al. Translational outpatient memory clinic working group. The new Alzheimer's criteria in a naturalistic series of patients with mild cognitive impairment. Journal of Neurology. 2010;257(12):2004–2014. doi: 10.1007/s00415-010-5650-0. [DOI] [PubMed] [Google Scholar]
  • 31.Kanai M, Matsubara E, Isoe K, et al. Longitudinal study of cerebrospinal fluid levels of tau, Aβ1-40, and Aβ1-42(43) in Alzheimer’s disease: a study in Japan. Annals of Neurology. 1998;44(1):17–26. doi: 10.1002/ana.410440108. [DOI] [PubMed] [Google Scholar]
  • 32.Mehta PD, Pirttilä T, Mehta SP, Sersen EA, Aisen PS, Wisniewski HM. Plasma and cerebrospinal fluid levels of amyloid β proteins 1-40 and 1- 42 in Alzheimer disease. Archives of Neurology. 2000;57(1):100–105. doi: 10.1001/archneur.57.1.100. [DOI] [PubMed] [Google Scholar]
  • 33.Fukuyama R, Mizuno T, Mizuno T, et al. Age-dependent change in the levels of Aβ40 and Aβ42 in cerebrospinal fluid from control subjects, and a decrease in the ratio of Aβ42 to Aβ40 level in cerebrospinal fluid from Alzheimer’s disease patients. European Neurology. 2000;43(3):155–160. doi: 10.1159/000008156. [DOI] [PubMed] [Google Scholar]
  • 34.Hansson O, Zetterberg H, Buchhave P, et al. Prediction of Alzheimer’s disease using the CSF Aβ42/Aβ40 ratio in patients with mild cognitive impairment. Dementia and Geriatric Cognitive Disorders. 2007;23(5):316–320. doi: 10.1159/000100926. [DOI] [PubMed] [Google Scholar]
  • 35.Fagan AM, Head D, Shah AR, et al. Decreased cerebrospinal fluid Aβ42 correlates with brain atrophy in cognitively normal elderly. Annals of Neurology. 2009;65(2):176–183. doi: 10.1002/ana.21559. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Roe CM, Fagan AM, Williams MM, et al. Improving CSF biomarker accuracy in predicting prevalent and incident Alzheimer disease. Neurology. 2011;76(6):501–510. doi: 10.1212/WNL.0b013e31820af900. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Rolstad S, Berg AI, Bjerke M, et al. Amyloid-β42 is associated with cognitive impairment in healthy elderly and subjective cognitive impairment. doi: 10.3233/JAD-2011-110038. Journal of Alzheimer’s Disease,. In press. [DOI] [PubMed] [Google Scholar]
  • 38.Cedazo-Minguez A, Winblad B. Biomarkers for Alzheimer’s disease and other forms of dementia: clinical needs, limitations and future aspects. Experimental Gerontology. 2010;45(1):5–14. doi: 10.1016/j.exger.2009.09.008. [DOI] [PubMed] [Google Scholar]
  • 39.Bibl M, Mollenhauer B, Wolf S, et al. Reduced CSF carboxyterminally truncated Aβ peptides in frontotemporal lobe degenerations. Journal of Neural Transmission. 2007;114(5):621–628. doi: 10.1007/s00702-006-0618-z. [DOI] [PubMed] [Google Scholar]
  • 40.Bibl M, Mollenhauer B, Esselmann H, et al. CSF diagnosis of Alzheimer’s disease and dementia with Lewy bodies. Journal of Neural Transmission. 2006;113(11):1771–1778. doi: 10.1007/s00702-006-0537-z. [DOI] [PubMed] [Google Scholar]
  • 41.Andreasen N, Zetterberg H. Amyloid-related biomarkers for Alzheimer’s disease. Current Medicinal Chemistry. 2008;15(8):766–771. doi: 10.2174/092986708783955572. [DOI] [PubMed] [Google Scholar]
  • 42.Gowing E, Roher AE, Woods AS, et al. Chemical characterization of Aβ 17-42 peptide, a component of diffuse amyloid deposits of Alzheimer disease. Journal of Biological Chemistry. 1994;269(15):10987–10990. [PubMed] [Google Scholar]
  • 43.Näslund J, Schierhorn A, Hellman U, et al. Relative abundance of Alzheimer Aβ amyloid peptide variants in Alzheimer disease and normal aging. Proceedings of the National Academy of Sciences of the United States of America. 1994;91(18):8378–8382. doi: 10.1073/pnas.91.18.8378. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Harigaya Y, Saido TC, Eckman CB, Prada CM, Shoji M, Younkin SG. Amyloid β protein starting pyroglutamate at position 3 is a major component of the amyloid deposits in the Alzheimer’s disease brain. Biochemical and Biophysical Research Communications. 2000;276(2):422–427. doi: 10.1006/bbrc.2000.3490. [DOI] [PubMed] [Google Scholar]
  • 45.Miravalle L, Calero M, Takao M, Roher AE, Ghetti B, Vidal R. Amino-terminally truncated Aβ peptide species are the main component of cotton wool plaques. Biochemistry. 2005;44(32):10810–10821. doi: 10.1021/bi0508237. [DOI] [PubMed] [Google Scholar]
  • 46.Piccini A, Russo C, Gliozzi A, et al. β-amyloid is different in normal aging and in Alzheimer disease. Journal of Biological Chemistry. 2005;280(40):34186–34192. doi: 10.1074/jbc.M501694200. [DOI] [PubMed] [Google Scholar]
  • 47.Portelius E, Westman-Brinkmalm A, Zetterberg H, Blennow K. Determination of β-amyloid peptide signatures in cerebrospinal fluid using immunoprecipitation-mass spectrometry. Journal of Proteome Research. 2006;5(4):1010–1016. doi: 10.1021/pr050475v. [DOI] [PubMed] [Google Scholar]
  • 48.Portelius E, Tran AJ, Andreasson U, et al. Characterization of amyloid β peptides in cerebrospinal fluid by an automated immunoprecipitation procedure followed by mass spectrometry. Journal of Proteome Research. 2007;6(11):4433–4439. doi: 10.1021/pr0703627. [DOI] [PubMed] [Google Scholar]
  • 49.Portelius E, Andreasson U, Ringman JM, et al. Distinct cerebrospinal fluid amyloid peptide signatures in sporadic and PSEN1 A431E-associated familial Alzheimer’s disease. Molecular Neurodegeneration. 2010;5(1, article 2) doi: 10.1186/1750-1326-5-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Maddalena AS, Papassotiropoulos A, Gonzalez-Agosti C, et al. Cerebrospinal fluid profile of amyloid β peptides in patients with Alzheimer’s disease determined by protein biochip technology. Neurodegenerative Diseases. 2004;1(4-5):231–235. doi: 10.1159/000080991. [DOI] [PubMed] [Google Scholar]
  • 51.Albertini V, Bruno A, Paterlini A, et al. Optimization protocol for amyloid-β peptides detection in human cerebrospinal fluid using SELDI TOF MS. Proteomics - Clinical Applications. 2010;4(3):352–357. doi: 10.1002/prca.200900166. [DOI] [PubMed] [Google Scholar]
  • 52.Portelius E, Zetterberg H, Andreasson U, et al. An Alzheimer’s disease-specific β-amyloid fragment signature in cerebrospinal fluid. Neuroscience Letters. 2006;409(3):215–219. doi: 10.1016/j.neulet.2006.09.044. [DOI] [PubMed] [Google Scholar]
  • 53.Vigo-Pelfrey C, Lee D, Keim P, Lieberburg I, Schenk DB. Characterization of β-amyloid peptide from human cerebrospinal fluid. Journal of Neurochemistry. 1993;61(5):1965–1968. doi: 10.1111/j.1471-4159.1993.tb09841.x. [DOI] [PubMed] [Google Scholar]
  • 54.Ghidoni R, Albertini V, Squitti R, et al. Novel T719P AβPP mutation unbalances the relative proportion of amyloid-β peptides. Journal of Alzheimer’s Disease. 2009;18(2):295–303. doi: 10.3233/JAD-2009-1142. [DOI] [PubMed] [Google Scholar]
  • 55.Lewczuk P, Esselmann H, Meyer M, et al. The amyloid-β (Aβ) peptide pattern in cerebrospinal fluid in Alzheimer’s disease: evidence of a novel carboxyterminally elongated Aβ peptide. Rapid Communications in Mass Spectrometry. 2003;17(12):1291–1296. doi: 10.1002/rcm.1048. [DOI] [PubMed] [Google Scholar]
  • 56.Vanderstichele H, De Meyer G, Andreasen N, et al. Amino-truncated β-amyloid42 peptides in cerebrospinal fluid and prediction of progression of mild cognitive impairment. Clinical Chemistry. 2005;51(9):1650–1660. doi: 10.1373/clinchem.2005.051201. [DOI] [PubMed] [Google Scholar]
  • 57.Wiltfang J, Esselmann H, Bibl M, et al. Highly conserved and disease-specific patterns of carboxyterminally truncated Aβ peptides 1-37/38/39 in addition to 1-40/42 in Alzheimer’s disease and in patients with chronic neuroinflammation. Journal of Neurochemistry. 2002;81(3):481–496. doi: 10.1046/j.1471-4159.2002.00818.x. [DOI] [PubMed] [Google Scholar]
  • 58.Lewczuk P, Esselmann H, Groemer TW, et al. Amyloid β peptides in cerebrospinal fluid as profiled with surface enhanced laser desorption/ionization time-of-flight mass spectrometry: evidence of novel biomarkers in Alzheimer’s disease. Biological Psychiatry. 2004;55(5):524–530. doi: 10.1016/j.biopsych.2003.10.014. [DOI] [PubMed] [Google Scholar]
  • 59.Portelius E, Dean RA, Gustavsson MK, et al. A novel abeta isoform pattern in CSF reflects gamma-secretase inhibition in Alzheimer disease. Alzheimer’s Research & Theraphy. 2010;2(7):1–7. doi: 10.1186/alzrt30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Walsh DM, Selkoe DJ. Aβ oligomers—a decade of discovery. Journal of Neurochemistry. 2007;101(5):1172–1184. doi: 10.1111/j.1471-4159.2006.04426.x. [DOI] [PubMed] [Google Scholar]
  • 61.Xia W, Yang T, Shankar G, et al. A specific enzyme-linked immunosorbent assay for measuring β-amyloid protein oligomers in human plasma and brain tissue of patients with Alzheimer Disease. Archives of Neurology. 2009;66(2):190–199. doi: 10.1001/archneurol.2008.565. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Lambert MP, Barlow AK, Chromy BA, et al. Diffusible, nonfibrillar ligands derived from Aβ1-42 are potent central nervous system neurotoxins. Proceedings of the National Academy of Sciences of the United States of America. 1998;95(11):6448–6453. doi: 10.1073/pnas.95.11.6448. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Hartley DM, Walsh DM, Ye CP, et al. Protofibrillar intermediates of amyloid β-protein induce acute electrophysiological changes and progressive neurotoxicity in cortical neurons. Journal of Neuroscience. 1999;19(20):8876–8884. doi: 10.1523/JNEUROSCI.19-20-08876.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Kayed R, Sokolov Y, Edmonds B, et al. Permeabilization of lipid bilayers is a common conformation-dependent activity of soluble amyloid oligomers in protein misfolding diseases. Journal of Biological Chemistry. 2004;279(45):46363–46366. doi: 10.1074/jbc.C400260200. [DOI] [PubMed] [Google Scholar]
  • 65.Mucke L, Masliah E, Yu GQ, et al. High-level neuronal expression of Aβ(1-42) in wild-type human amyloid protein precursor transgenic mice: synaptotoxicity without plaque formation. Journal of Neuroscience. 2000;20(11):4050–4058. doi: 10.1523/JNEUROSCI.20-11-04050.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Lesné S, Ming TK, Kotilinek L, et al. A specific amyloid-β protein assembly in the brain impairs memory. Nature. 2006;440(7082):352–357. doi: 10.1038/nature04533. [DOI] [PubMed] [Google Scholar]
  • 67.Walsh DM, Klyubin I, Fadeeva JV, et al. Naturally secreted oligomers of amyloid β protein potently inhibit hippocampal long-term potentiation in vivo. Nature. 2002;416(6880):535–539. doi: 10.1038/416535a. [DOI] [PubMed] [Google Scholar]
  • 68.Klyubin I, Walsh DM, Lemere CA, et al. Amyloid β protein immunotherapy neutralizes Aβ oligomers that disrupt synaptic plasticity in vivo. Nature Medicine. 2005;11(5):556–561. doi: 10.1038/nm1234. [DOI] [PubMed] [Google Scholar]
  • 69.Shankar GM, Li S, Mehta TH, et al. Amyloid-β protein dimers isolated directly from Alzheimer’s brains impair synaptic plasticity and memory. Nature Medicine. 2008;14(8):837–842. doi: 10.1038/nm1782. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Reed MN, Hofmeister JJ, Jungbauer L, et al. Cognitive effects of cell-derived and synthetically derived Abeta oligomers. Neurobiology of Aging. 2011;32(10):1784–1794. doi: 10.1016/j.neurobiolaging.2009.11.007. Article ID S0197-4580(09)00367-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Li S, Jin M, Koeglsperger T, Shepardson NE, Shankar GM, Selkoe DJ. Soluble a β oligomers inhibit long-term potentiation through a mechanism involving excessive activation of extrasynaptic NR2B-containing NMDA receptors. Journal of Neuroscience. 2011;31(18):6627–6638. doi: 10.1523/JNEUROSCI.0203-11.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Pitschke M, Prior R, Haupt M, Riesner D. Detection of single amyloid β-protein aggregates in the cerebrospinal fluid of Alzheirner’s patients by fluorescence correlation spectroscopy. Nature Medicine. 1998;4(7):832–834. doi: 10.1038/nm0798-832. [DOI] [PubMed] [Google Scholar]
  • 73.Georganopoulou DG, Chang L, Nam JM, et al. Nanoparticle-based detection in cerebral spinal fluid of a soluble pathogenic biomarker for Alzheimer’s disease. Proceedings of the National Academy of Sciences of the United States of America. 2005;102(7):2273–2276. doi: 10.1073/pnas.0409336102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Santos AN, Torkler S, Nowak D, et al. Detection of amyloid-β oligomers in human cerebrospinal fluid by flow cytometry and fluorescence resonance energy transfer. Journal of Alzheimer’s Disease. 2007;11(1):117–125. doi: 10.3233/jad-2007-11114. [DOI] [PubMed] [Google Scholar]
  • 75.Klyubin I, Betts V, Welzel AT, et al. Amyloid β protein dimer-containing human CSF disrupts synaptic plasticity: prevention by systemic passive immunization. Journal of Neuroscience. 2008;28(16):4231–4237. doi: 10.1523/JNEUROSCI.5161-07.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Englund H, Gunnarsson MD, Brundin RM, et al. Oligomerization partially explains the lowering of Aβ42 in alzheimer’s disease cerebrospinal fluid. Neurodegenerative Diseases. 2009;6(4):139–147. doi: 10.1159/000225376. [DOI] [PubMed] [Google Scholar]
  • 77.Fukumoto H, Tokuda T, Kasai T, et al. High-molecular-weight beta-amyloid oligomers are elevated in cerebrospinal fluid of Alzheimer patients. The Journal the Federation of American Societies for Experimental Biology. 2010;24(8):2716–2726. doi: 10.1096/fj.09-150359. [DOI] [PubMed] [Google Scholar]
  • 78.Gao CM, Yam AY, Magdangal E, et al. Aβ40 oligomers identified as a potential biomarker for the diagnosis of alzheimer's disease. PLoS One. 2010;5(12) doi: 10.1371/journal.pone.0015725. Article ID e15725. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Kayed R, Head E, Thompson JL, et al. Common structure of soluble amyloid oligomers implies common mechanism of pathogenesis. Science. 2003;300(5618):486–489. doi: 10.1126/science.1079469. [DOI] [PubMed] [Google Scholar]
  • 80.O’Nuallain B, Wetzel R. Conformational Abs recognizing a generic amyloid fibril epitope. Proceedings of the National Academy of Sciences of the United States of America. 2002;99(3):1485–1490. doi: 10.1073/pnas.022662599. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Lambert MP, Velasco PT, Chang L, et al. Monoclonal antibodies that target pathological assemblies of Aβ. Journal of Neurochemistry. 2007;100(1):23–35. doi: 10.1111/j.1471-4159.2006.04157.x. [DOI] [PubMed] [Google Scholar]
  • 82.Habicht G, Haupt C, Friedrich RP, et al. Directed selection of a conformational antibody domain that prevents mature amyloid fibril formation by stabilizing Aβ protofibrils. Proceedings of the National Academy of Sciences of the United States of America. 2007;104(49):19232–19237. doi: 10.1073/pnas.0703793104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Meli G, Visintin M, Cannistraci I, Cattaneo A. Direct in vivo intracellular selection of conformation-sensitive antibody domains targeting Alzheimer’s amyloid-β oligomers. Journal of Molecular Biology. 2009;387(3):584–606. doi: 10.1016/j.jmb.2009.01.061. [DOI] [PubMed] [Google Scholar]
  • 84.Agnati LF, Leo G, Genedani S, et al. Common key-signals in learning and neurodegeneration: focus on excito-amino acids, β-amyloid peptides and α-synuclein. Journal of Neural Transmission. 2009;116(8):953–974. doi: 10.1007/s00702-008-0150-4. [DOI] [PubMed] [Google Scholar]
  • 85.Agnati LF, Fuxe K. Volume transmission as a key feature of information handling in the central nervous system possible new interpretative value of the Turing’s B-type machine. Progress in Brain Research. 2000;125:3–19. doi: 10.1016/S0079-6123(00)25003-6. [DOI] [PubMed] [Google Scholar]
  • 86.Lakkaraju A, Rodriguez-Boulan E. Itinerant exosomes: emerging roles in cell and tissue polarity. Trends in Cell Biology. 2008;18(5):199–209. doi: 10.1016/j.tcb.2008.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Cocucci E, Racchetti G, Meldolesi J. Shedding microvesicles: artefacts no more. Trends in Cell Biology. 2009;19(2):43–51. doi: 10.1016/j.tcb.2008.11.003. [DOI] [PubMed] [Google Scholar]
  • 88.Simons M, Raposo G. Exosomes—vesicular carriers for intercellular communication. Current Opinion in Cell Biology. 2009;21(4):575–581. doi: 10.1016/j.ceb.2009.03.007. [DOI] [PubMed] [Google Scholar]
  • 89.Rustom A. Hen or egg? Some thoughts on tunneling nanotubes. Annals of the New York Academy of Sciences. 2009;1178:129–136. doi: 10.1111/j.1749-6632.2009.04997.x. [DOI] [PubMed] [Google Scholar]
  • 90.Agnati LF, Genedani S, Leo G, et al. Aβ peptides as one of the crucial volume transmission signals in the trophic units and their interactions with homocysteine. Physiological implications and relevance for Alzheimer’s disease. Journal of Neural Transmission. 2007;114(1):21–31. doi: 10.1007/s00702-006-0564-9. [DOI] [PubMed] [Google Scholar]
  • 91.Vekrellis K, Ye Z, Qiu WQ, et al. Neurons regulate extracellular levels of amyloid β-protein via proteolysis by insulin-degrading enzyme. Journal of Neuroscience. 2000;20(5):1657–1665. doi: 10.1523/JNEUROSCI.20-05-01657.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Hock C, Golombowski S, Müller-Spahn F, et al. Cerebrospinal fluid levels of amyloid precursor protein and amyloid β-peptide in Alzheimer’s disease and major depression—inverse correlation with dementia severity. European Neurology. 1998;39(2):111–118. doi: 10.1159/000007917. [DOI] [PubMed] [Google Scholar]
  • 93.Funke SA, Birkmann E, Willbold D. Detection of amyloid-β aggregates in body fluids: a suitable method for early diagnosis of Alzheimer’s disease? Current Alzheimer Research. 2009;6(3):285–289. doi: 10.2174/156720509788486536. [DOI] [PubMed] [Google Scholar]
  • 94.Oe T, Ackermann BL, Inoue K, et al. Quantitative analysis of amyloid β peptides in cerebrospinal fluid of Alzheimer’s disease patients by immunoaffinity purification and stable isotope dilution liquid chromatography/negative electrospray ionization tandem mass spectrometry. Rapid Communications in Mass Spectrometry. 2006;20(24):3723–3735. doi: 10.1002/rcm.2787. [DOI] [PubMed] [Google Scholar]
  • 95.Rajendran L, Honsho M, Zahn TR, et al. Alzheimer’s disease β-amyloid peptides are released in association with exosomes. Proceedings of the National Academy of Sciences of the United States of America. 2006;103(30):11172–11177. doi: 10.1073/pnas.0603838103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Vingtdeux V, Hamdane M, Loyens A, et al. Alkalizing drugs induce accumulation of amyloid precursor protein by-products in luminal vesicles of multivesicular bodies. Journal of Biological Chemistry. 2007;282(25):18197–18205. doi: 10.1074/jbc.M609475200. [DOI] [PubMed] [Google Scholar]
  • 97.Sharples RA, Vella LJ, Nisbet RM, et al. Inhibition of γ-secretase causes increased secretion of amyloid precursor protein C-terminal fragments in association with exosomes. The Journal of the Federation of American Societies for Experimental Biology. 2008;22(5):1469–1478. doi: 10.1096/fj.07-9357com. [DOI] [PubMed] [Google Scholar]
  • 98.Ghidoni R, Benussi L, Binetti G. Exosomes: the Trojan horses of neurodegeneration. Medical Hypotheses. 2008;70(6):1226–1227. doi: 10.1016/j.mehy.2007.12.003. [DOI] [PubMed] [Google Scholar]
  • 99.Ghidoni R, Paterlini A, Albertini V, et al. Cystatin C is released in association with exosomes: a new tool of neuronal communication which is unbalanced in Alzheimer’s disease. Neurobiology of Aging. 2009;32(8):1435–1442. doi: 10.1016/j.neurobiolaging.2009.08.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Agnati LF, Guidolin D, Baluka F, et al. A new hypothesis of pathogenesis based on the divorce between mitochondria and their host cells: possible relevance for alzheimer’s disease. Current Alzheimer Research. 2010;7(4):307–322. doi: 10.2174/156720510791162395. [DOI] [PubMed] [Google Scholar]
  • 101.Chi EY, Frey SL, Lee KYC. Ganglioside GM1-mediated amyloid-beta fibrillogenesis and membrane disruption. Biochemistry. 2007;46(7):1913–1924. doi: 10.1021/bi062177x. [DOI] [PubMed] [Google Scholar]
  • 102.Yuyama K, Yamamoto N, Yanagisawa K. Accelerated release of exosome-associated GM1 ganglioside (GM1) by endocytic pathway abnormality: another putative pathway for GM1-induced amyloid fibril formation. Journal of Neurochemistry. 2008;105(1):217–224. doi: 10.1111/j.1471-4159.2007.05128.x. [DOI] [PubMed] [Google Scholar]
  • 103.Re RN, Cook JL. Senescence, apoptosis, and stem cell biology: the rationale for an expanded view of intracrine action. American Journal of Physiology, Heart and Circulatory Physiology. 2009;297(3):H893–H901. doi: 10.1152/ajpheart.00414.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Lai AY, McLaurin J. Mechanisms of amyloid-beta peptide uptake by neurons: the role of lipid rafts and lipid raft-associated proteins. International Journal of Alzheimer's Disease. 2011;2011:11 pages. doi: 10.4061/2011/548380. Article ID 548380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Pearson HA, Peers C. Physiological roles for amyloid β peptides. Journal of Physiology. 2006;575(1):5–10. doi: 10.1113/jphysiol.2006.111203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Garcia-Osta A, Alberini CM. Amyloid beta mediates memory formation. Neurobiology of Learning and Memory. 2009;16(4):267–272. doi: 10.1101/lm.1310209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Soscia SJ, Kirby JE, Washicosky KJ, et al. The Alzheimer’s disease-associated amyloid β-protein is an antimicrobial peptide. PLoS One. 2010;5(3):1–10. doi: 10.1371/journal.pone.0009505. Article ID e9505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Tanzi E. I fatti e le induzione nell’odierna istologia del sistema nervoso. Rivista sperimentale di freniatria e medicina legale delle alienazioni mentali. 1893;19:419–472. [Google Scholar]
  • 109.Taylor JS, Gaze RM. The effects of the fibre environment on the paths taken by regenerating optic nerve fibres in Xenopus. Journal of Embryology and Experimental Morphology. 1985;89:383–401. [PubMed] [Google Scholar]
  • 110.Agnati LF, Zoli M, Biagini G, Fuxe K. Neuronal plasticity and ageing processes in the frame of the ’Red Queen Theory’. Acta Physiologica Scandinavica. 1992;145(4):301–309. doi: 10.1111/j.1748-1716.1992.tb09370.x. [DOI] [PubMed] [Google Scholar]
  • 111.Yamada K, Nabeshima T. Brain-derived neurotrophic factor/TrkB signaling in memory processes. Journal Pharmacological Sciences. 2003;91(4):267–270. doi: 10.1254/jphs.91.267. [DOI] [PubMed] [Google Scholar]
  • 112.Schindowski K, Belarbi K, Buée L. Neurotrophic factors in Alzheimer’s disease: role of axonal transport. Genes, Brain and Behavior. 2008;7(1):43–56. doi: 10.1111/j.1601-183X.2007.00378.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Tuppo EE, Arias HR. The role of inflammation in Alzheimer’s disease. International Journal of Biochemistry and Cell Biology. 2005;37(2):289–305. doi: 10.1016/j.biocel.2004.07.009. [DOI] [PubMed] [Google Scholar]

Articles from Journal of Biomedicine and Biotechnology are provided here courtesy of Wiley

RESOURCES