Skip to main content
  • Review
  • Published:

Relationships between the amyloid precursor protein and its various proteolytic fragments and neuronal systems

Abstract

Alzheimer's disease (AD) is a progressive neurodegenerative disease and in its familial form is associated with mutations in the amyloid precursor protein (APP) and the presenilins (PSs). Much data regarding the interactions of APP, its proteolytic fragments and PS have been generated, expanding our understanding of the roles of these proteins in mechanisms underlying cognitive function and revealing many complex relationships with wide ranging cellular systems. In this review, we examine the multiple interactions of APP and its proteolytic fragments with other neuronal systems in terms of feedback loops and use these relationships to build a map. We highlight the complexity involved in the APP proteolytic system and discuss alternative perspectives on the roles of APP and its proteolytic fragments in dynamic processes associated with disease progression in AD. We highlight areas where data are missing and suggest potential confounding factors. We suggest that a systems biology approach enhances representations of the data and may be more useful in modelling both normal cognition and disease processes.

Introduction

Alzheimer's disease (AD) is a clinico-pathological diagnosis in which progressive neurodegenerative disease is associated with the dementia syndrome. Neuropathologically, AD is generally characterised as including deposits of amyloid beta protein (Aβ) in senile and neuritic plaques [1] and neurofibrillary tangles of paired helical filaments of hyperphosphorylated tau [2, 3]. Currently, AD is classified as familial AD, which has an early age of onset and is associated with mutations in either the amyloid precursor protein (APP) or the presenilins (PSs), and sporadic AD, which has a later age of onset and a less clear aetiology [4, 5]. Down's syndrome, with an extra copy of APP on chromosome 21, also leads to a form of AD with increasing age. The genetic data associated with familial AD [6] strongly suggest that PS, APP and its proteolytic fragment Aβ are involved in AD disease progression but exactly how they contribute to both normal cognition and dementia is the focus of much debate.

Many non-genetic factors also appear to affect disease initiation and progression, including diet, exercise and education, and many models have been proposed to account for their roles in AD. Population studies (for example, the Medical Research Council Cognitive Function and Aging Study [7]) demonstrate that vascular and AD-associated pathologies are common in people with and without any clinical manifestation of dementia. Coupled with data suggesting that the relationship between amyloid pathology and dementia attenuates with age [8], this suggests that sporadic AD especially has a complex disease aetiology [9].

Many models of disease progression have been proposed to account for the accumulating data on AD. These models form the basis of experimental design and affect how findings are interpreted. Data relevant to disease progression in AD are accumulating from many diverse fields, including neuropsychology, brain imaging, molecular biology and genetics. Integrating these data is becoming harder as expertise is required in so many areas.

The pre-eminent model of the last few decades has been the amyloid cascade hypothesis [10–12], based on genetic data and the role of PS in the proteolysis of APP to release the 39 to 43 residue Aβ. This model proposes that overproduction of Aβ, or an increase in Aβ(1-42) relative to Aβ(1-40), is causal in the disease process. In this model, Aβ, perhaps in a specific aggregation state, is proposed as neurotoxic. An alternative interpretation of data relating to APP, PS and Aβ is the presenilin hypothesis [13], where loss or altered function of PS is proposed as the causal factor in disease progression. Other models suggest different initiating factors, such as cholesterol [14], mitochondria and apoptosis [15], insulin signalling and energy metabolism [16, 17], cholinergic insufficiency [18], Ca2+ deregulation [19, 20], signalling decoherence [21] or a combination of multiple factors (for example, [22, 23]). No single model adequately explains how APP, PS, cholesterol and other factors interact with synaptic plasticity, learning and memory in normal and disease states. This suggests that models currently in use in AD research require re-evaluation.

Recent approaches to integrating the data relevant to AD have involved the use of text mining, bioinformatic databases and network analysis for gene co-expression and protein-protein interactions [24–33]. Studies using automated interrogation of genomic and proteomic databases, such as UNIPROTKB [34], NCBI Entrez Gene [35], and IntAct [36], and text mining in literature databases such as Pubmed [37] have confirmed the involvement of many previously highlighted neurological processes, extended the connectivity between known molecular targets and revealed new targets for investigation (summarised in Table 1).

Table 1 Overview of protein-protein interaction network studies

Understanding how APP contributes to synaptic plasticity is important in understanding normal cognition and disease progression in AD. Here, we approach the process of integrating the available data relating to the relationships between APP and its proteolytic fragments and neuronal processes based on a systems biology approach and build a map of relationships. We are not aiming to define protein interactions nor find new targets for investigation, but to highlight relationships in the existing literature that illustrate the complexity involved and how this relates to our current understanding of the role of APP in AD. We highlight areas where data are missing and predict relationships that may confound current disease models.

Building the map

Relationships involving APP and each of its proteolytic fragments were identified from literature searches using relevant key words. Each interaction identified was manually entered into a non-hierarchical network using the mapping software Compendium [38]. This software allowed each molecule or neuronal system to be represented as a node within the map. Each node in the map could contain further networks of nodes, leading to a multi-dimensional organisation of information. These further nodes represented information relevant to the molecule of the parent node and could include notes describing possible relevance, questions raised and links to relevant entries on various bioinformatic databases. The map is not meant to be read as a protein-protein interaction network (PPI), but rather was designed to summarise and collate information relevant to the APP system in a more general way. Physiological outcomes, such as changes in signalling, were included, even though the precise molecular interactions are not clear. Any additional details, such as new biological relationships or new molecular interactions, can be added as they arise. Relationships between molecules are represented by connectors, with annotation if required. Figure 1 shows a limited map built in this way from references cited in the text, with additional references listed in Table 2.

Figure 1
figure 1

A limited map of amyloid precursor protein relationships showing molecules/neuronal systems as nodes and interactions between them as arrows. Evidence for each relationship is summarised in Table 2. Numbers are referred to in the text. Aβ, amyloid beta protein; Abl, tyrosine-protein kinase ABL1; AChR, acetylcholine receptor; ADAM, a disintegrin and metalloproteinase domain-containing protein; AICD, APP intracellular domain; Akt, RAC-alpha serine/threonine-protein kinase; AMPAR, AMPA receptor; ApoE, apolipoprotein E; APP, amyloid precursor protein; BACE, beta-site amyloid precursor protein cleaving enzyme; CD74, HLA class II histocompatibility antigen gamma chain; ECM, extracellular matrix; IDE, insulin degrading enzyme; JIP1, C-Jun-amino-terminal kinase-interacting protein 1; JNK, C-Jun amino-terminal kinase; Lyn, tyrosine-protein kinase Lyn; PKA, protein kinase A; PKC, protein kinase C; LRP, low-density lipoprotein receptor; LTP, long-term potentiation; mAChR, muscarinic acetylcholine receptor; mGluR, metabotropic glutamate receptor; nAChR, nicotinic acetylcholine receptor; NMDAR, NMDA receptor; P2Y2, P2Y purinoceptor 2; PI3K, phosphoinositide 3-kinase; sAPP, secreted amyloid precursor protein; Src, proto-oncogene tyrosine-protein kinase Src; TIMP, tissue inhibitor of metalloproteinases; TNF, tumour necrosis factor.

Table 2 Additional references supporting the network diagram in Figure 1

Unlike more automated network studies such as that by Perreau and colleagues [25] where nodes were included in the network only if there was direct evidence of molecular interaction, or various studies [30–33] where co-expression criteria were applied, no specific criteria were imposed and any relationship found was included. Known differences in peptide behaviour (for example, [39, 40]) necessitated separate nodes for Aβ(1-40) and Aβ(1-42). This was extended systematically to include all the peptide fragments, including P3(17-40), P3(17-42), secreted APP (sAPP)α and sAPPβ (the extracellular amino-terminal fragments following α- and β-cleavage, respectively), C31, and so on to take account of any potential behavioural differences. This is in contrast to various network constructions where the various proteolytic fragments are collapsed into an APP parental node, possibly reflecting the UNIPROTKB identifier (P05067) being the same for each peptide. Additionally, different aggregation states of each of the peptides (monomers, dimers, oligomers or fibrils) were considered separately. These additional details have been omitted from Figure 1 for clarity. Since there was little or no evidence for many of the Aβ-type peptides [41], these were collapsed into a separate node (not shown).

The relationships in Figure 1 were analysed manually by identifying feedback loops. Direct feedback loops were defined as relationships between APP, any of its proteolytic fragments and any other molecule or functional change that was represented at more than one point in the map. Possible indirect feedback loops between any neural system, such as neurotransmitter signalling, Ca2+ homeostasis or the inflammation cascade, were also investigated by looking for molecules or functional change in these systems that interact with APP or any of its proteolytic fragments at more than one point in the map.

Following a brief summary of APP cleavage, two examples of the relationships between APP pathways and neuronal processes, looking at the extracellular matrix (ECM) and endocytosis, serve to illustrate the implications of the complexity involved.

The amyloid precursor protein

Overview

APP is a type I transmembrane protein [42] with a large extracellular amino-terminal domain, a transmembrane domain and an intracellular carboxy-terminal domain that resembles a cell surface receptor [43]. It is expressed in several isoforms, with APP695 the main isoform expressed by neurons. APP has a high turnover, with a half life ranging from approximately 1 hour [44–46] to approximately 4 hours [47–49]. In addition to its functions at the cell surface, full length mature APP is also processed via competing pathways that release proteolytic fragments [5, 43, 50] (recently reviewed in [51]). In effect there are four possible routes: APP may remain functionally active at the cell surface; it may be internalised and recycled or degraded; it may be cleaved via the α-pathway, releasing the sAPPα fragment, leaving the membrane bound C83 carboxy-terminal fragment that is further processed to the P3 fragment; or it may be cleaved via the β-pathway releasing sAPPβ, leaving the membrane bound C99 carboxy-terminal fragment that is further processed to the Aβ fragment.

As can be seen in Figure 1, α- and β-cleavage pathways (numbered 1 and 2, respectively) converge on a further intramembrane γ-cleavage (numbered 3) by the PS-containing γ-secretase complex to release the variable length Aβ and P3 peptides and the variable length APP intracellular domain (AICD). Additional cleavages, including caspase cleavage of the carboxyl terminus at residue D664 of APP695, producing the alternative length carboxy-terminal fragment C31 [51], and cleavage by BACE2 (beta-site amyloid precursor protein cleaving enzyme 2) within the Aβ sequence [52], have been omitted for clarity.

Unprocessed APP is degraded or recycled via the endosomal or lysosomal pathways and may be recycled back to the membrane and processed within approximately 30 minutes [46], with perhaps a third to a half being processed via the cleavage pathways as measured by secreted sAPPα/β [46]. Regulatory relationships promoting expression or cleavage have been collapsed into summary boxes in Figure 1 for simplicity.

Complex feedback pathways involving APP and its proteolytic fragments; the extracellular matrix and endocytosis

The extracellular matrix

APP interacts with itself and its own proteolytic fragments at many points in the network with various functional consequences. At the cell surface, the large amino-terminal domain of full length APP interacts with many components of the ECM, including heparins and heparin sulphate proteoglycans [42], laminin, collagen [53, 54] and β-1-integrin [43, 55, 56], and contributes to early neuronal development, axon and neurite growth, cell adhesion and synaptic plasticity [43, 57]. Many of these interactions are now represented in MatrixDB [58], an ECM interaction database.

The amino-terminal domain of APP binds heparin in a pH- and zinc-dependant manner and this binding promotes the formation of APP homodimers [42, 59–61]. Depending on various factors, including cell compartment and pH, this dimerisation may influence APP proteolysis via α- or β-cleavages due to conformational changes [59], though this requires further investigation.

The large amino-terminal fragment released by α-cleavage, sAPPα, is a soluble monomer. It retains two heparin binding sites and has been shown to bind heparin as a dimer [60]. The ability of sAPPα to disrupt APP dimerisation at the cell surface is thought to contribute to its neuroprotective actions [62–64] and may partly explain why sAPPα is approximately 100 times more neuroprotective against excitotoxicity, glucose deprivation and the addition of Aβ in hippocampal cultures than sAPPβ, which lacks the second carboxy-terminal heparin binding site [63]. Dementia status has been associated with both reduced sAPPα levels in cerebrospinal fluid [65] and an increased half life of sAPPα [49].

Differing by only 16 carboxy-terminal residues, sAPPα and sAPPβ share a high degree of sequence homology and have the potential to compete with each other for binding sites. Since both sAPPβ and sAPPα are constantly present in the ECM in dynamically changing ratios, we predict that sAPPβ could act as an antagonist of sAPPα/APP (shown in Figure 2 as competitive inhibition (line 1)), with the potential to modulate various interactions between APP and sAPPα and the ECM. We predict, therefore, that any functional outcome from the interactions of the APP pathway will depend on the synergy between APP and its amino-terminal fragments.

Figure 2
figure 2

Feedback between amyloid precursor protein and its proteolytic fragments. Inhibition is shown in red, enhancement in green; numbers are referred to in the text. Functionally different aggregation states for the Aβ-type peptides are collapsed into P3 and Aβ nodes for clarity. Aβ, amyloid beta protein; AICD, APP intracellular domain; APP, amyloid precursor protein; PKC, protein kinase C; sAPP, secreted amyloid precursor protein.

Aβ interacts with heparins in the ECM and at high levels may prevent the catabolism of proteoglycans and promote amyloid formation [66]. Reciprocally, heparins modulate many of the interactions involving Aβ. Heparins enhance both nucleation and elongation processes in the aggregation of Aβ [67] and modulate interactions of Aβ; for example, heparins can limit the neurotoxic and pro-inflammatory activity of Aβ in a dose-dependent manner [68]. Additionally, heparins contribute to the uptake of Aβ by a pathway shared with apolipoprotein E [69] and also contribute to the regulation of the β-secretase BACE in a dose-dependent manner, with low concentrations promoting and high concentrations inhibiting the activation of BACE1 [70].

In terms of direct feedback, Aβ may be part of a negative feedback loop involved in the regulation of APP expression and the promotion of α-cleavage. Fibrillar forms of various proteins, including Aβ, interact with amino-terminal APP and increase APP expression [71] (Figure 2, line 2) and Aβ(1-40) may up-regulate expression of α-secretase via integrin receptors and matrix metallo-proteinase-9 activity [72] (Figure 2, line 3). Additionally, Aβ has many indirect negative feedback relationships with α-cleavage - for example, Aβ inhibits protein kinase C [73] and insulin signalling [74], both involved in up-regulation of α-cleavage [75] (Figure 2, line 5) - and additionally may negatively feedback on APP expression and its own production via a pathway involving Fe65 and glycogen synthase kinase 3 [76]. Reciprocally, changes to gene expression by sAPPα via the activation of NF-κβ may attenuate the interactions of Aβ [77] and this can be understood as reciprocal indirect feedback from the α-pathway on the β-pathway (Figure 2, line 4).

The data reviewed above suggest that Aβ may contribute to both direct and indirect feedback loops in the ECM at multiple points, with consequences for the interactions of APP, sAPPα, Aβ and sAPPβ that may ultimately affect cell adhesion, neurite outgrowth and synaptic plasticity. These effects will depend on the specific sequence length, concentration and aggregation state of the Aβ-type peptides, which may dynamically change in response to the neuronal environment.

Endocytosis

Increases in endocytosis, often associated with synaptic activity, lead to both an increase in Aβ [78] and a reduction in α-cleavage and lower secreted sAPPα levels [79]. This may reflect the regulatory role of compartmentation in APP proteolysis, where α-cleavage occurs at the cell surface and Aβ production is associated with factors that promote endocytosis. No studies could be found investigating the role of endocytosis on P3 production, so this cannot be discussed specifically. Endocytosis allows a degree of dissociation between the α- and β-cleavages and γ-cleavage, resulting in levels of sAPPα and sAPPβ that may be independent of Aβ(1-40/42) and P3 levels, respectively. This means that in terms of function, APP processing via the β-pathway has the potential to either: modulate actions of sAPPα via competition with sAPPβ; or modulate the actions of sAPPα and initiate actions mediated by Aβ.

P3 corresponds to Aβ(17-40/42) and we predict that this shared sequence could allow P3 to modulate the interactions of Aβ (shown in Figure 2 as competitive inhibition), especially where those interactions involve the carboxy-terminal amino acids. While no studies can be found that address this particular question directly, various amino-terminal truncated forms of Aβ have been associated with diffuse plaques [80, 81] and the Aβ(17-40) fragment, corresponding to P3, has been studied in terms of aggregation [82]. It is clear that amino-truncated Aβ peptides, including P3, have potential for aggregation and can interact with Aβ, but how this affects the physiological behaviour of neuronal systems is not clear. In terms of functional flow through the APP pathway, processing via the α-pathway to release sAPPα and P3 could either: initiate actions of sAPPα; or initiate actions of sAPPα and modulate actions mediated by Aβ.

In terms of modelling these interactions in functional contexts, PPI networks may require a more detailed approach, where each sequence length and aggregation state is represented as an individual node.

The effects of concentration, sequence length aggregation state and affinity

Aβ is produced in a range of sequence lengths [41] and can form monomers, dimers, oligomers and fibrils. The amount of Aβ produced is regulated and can dynamically change in response to many factors, including increased cholesterol [83], increased synaptic activity [84], heparins via activation of BACE1 [70], reduced acetylcholine (Ach) signalling via muscarinic receptors [70], sustained increased cytosolic Ca2+ [85], and hypoxia [86]. Changes in concentration are a well recognised mechanism of regulation in cellular processes; for example, Aβ(1-40) and Aβ(1-42) both promote angiogenesis at nanomolar concentrations but inhibit it at higher micromolar concentrations [87].

Different sequence lengths show different behaviours; for example, Aβ(1-42) is more prone to aggregation than Aβ(1-40) due to a more rigid carboxyl terminus [88]. Aβ monomers appear to have structured and unstructured regions, with differences in aqueous conformations between Aβ(1-40) and Aβ(1-42) [89]. While both Aβ(1-40) and Aβ(1-42) reduced currents via α7 and α4β2 ACh receptors (AChRs), only Aβ(1-40) increased glutamatergic signalling via AMPA receptors [40]. Another study found that the effects of Aβ(1-40) and Aβ(1-42) on α7 AChRs, as measured by ACh release and Ca2+ influx, were different [39]. Additionally, Aβ(1-40) may protect neurons from the effects of Aβ(1-42) by disrupting the aggregation of Aβ(1-42) [90] and this raises the possibility that P3-type peptides may act in similar ways. The functional effects of sequence length are only just beginning to be elucidated and should be expanded to include all the Aβ-type peptides, including P3. In addition to differences in peptide sequence and concentration, aggregation state also modulates function. Aβ oligomers appear to be spherical [91, 92] and Aβ fibrils appear as fine fibres [92]. Aβ oligomers, but not monomers or fibrils, enhance lipid release [93] and at low concentrations enhance neuronal differentiation [92] whereas Aβ fibrils, but not monomers or oligomers, may interact with APP and increase its expression [71]. While both soluble and aggregated forms of Aβ(1-40) inhibit long-term potentiation, the fibrillar forms may additionally affect neurotransmitter release [94].

Within a specific cellular compartment, whether one interaction is more likely to occur than another depends on the relative affinities of the reactants involved. While dissociation constants have been measured for some reactions in rodent models (for example, K(i) values of Aβ(1-42) for α7 nicotinic AChRs from rat and guinea pig are 4.1 and 5.0 pM, respectively, and the K(i) of Aβ(1-42) with α4β2 AChRs is approximately 5,000-fold lower at 30 and 23 nM, respectively [95]), we are missing dissociation constants for most interactions in humans.

Taken together, the data discussed above suggest that the interactions of APP and its proteolytic fragments with synaptic systems are complex and involve subtle changes in the relative ratios between APP and all its fragments. Given the complexity of the APP system, multiple neuronal systems have the potential to regulate the expression and proteolysis of APP and could also initiate imbalance within this system, leading to multiple possible disease pathways.

This complexity has the potential to confound purely empirical studies into the behaviour of the peptides in model systems. Over-expressing Aβ peptides in, for example, mouse models without knowing their concentrations, sequences and aggregation states and the background concentrations, sequences and aggregation states of the other relevant peptides in the experimental system that is being studied means that we do not know if the experimental results obtained are due to the properties of the Aβ peptides themselves or reflect a disrupted balance in a complex system over time, or perhaps both. While this distinction may seem subtle, given the self-organising and self-referencing properties of the human brain, an approach based on the principles in systems biology may better represent the roles of the APP pathway in functional contexts.

Discussion

It is not possible to include the full extent of the known interaction of APP and its proteolytic fragments here and further interactions may yet be discovered. Data relating to the roles of APP and its proteolytic fragments in synaptic plasticity and AD have accumulated from many diverse fields and these need to be placed into context. One way of doing this is to investigate the relationships between APP, its proteolytic fragments and wider neuronal systems by mapping networks of interactions.

It is interesting that the networks generated in the various studies reviewed in Table 1 do not always overlap and different studies highlight different pathways or biological processes, for example, Fe2+ [28], apoptosis [96], or cardiovascular disease/diabetes [31]. Each study has different starting points, inclusion/exclusion criteria and network construction methods, so this lack of agreement is no surprise. It is difficult to assess the degree to which the various starting points, criteria and network construction methods bias results towards an outcome.

The study by Soler-Lopez and colleagues [33] may not model the interactions of full length APP in the membrane adequately, as many of the ECM proteins that might be expected to interact are excluded due to difficulties involved in expressing them in the experimental microarray used. This may shift the focus of their network more towards intracellular interactions. Given the importance of the various interactions of APP with components of the ECM, any study excluding such proteins and proteoglycans could be seriously confounded.

While the study by Perreau and colleagues [25] maps PPIs to specific domains of full length APP, they do not distinguish between the different proteolytic peptides. Additionally, transient interactions and biologically essential post-translational modifications, such as glycosylation and phosphorylation, cannot yet be fully represented in PPI networks [27], severely limiting the modelling of regulation and control in these models.

Studies comparing mouse and human gene expression networks [32] or aging and AD pathways [30] have revealed interesting results. Perhaps most striking is the study revealing a human-specific network for PS function in oligodendrocytes and myelination, with the potential to confound the current approach to modelling AD in rodents [32]. Miller and colleagues [30, 32] also show important contributions from both neuronal and glial pathways, perhaps reflecting the contributions of glia to disease pathways found in humans [97, 98]. The differences found in networks between glia and neurons could be extended to differentiate between specific neuronal types and future network models may usefully investigate why cholinergic neurons in AD appear more susceptible to degeneration than others.

We do not yet have totally inclusive, gene co-expression or PPI models of APP and related pathways to the level of detail required and this places limitations on their interpretation. Given the limitations, however, network studies do show the usefulness of the systems biology approach in integrating huge volumes of data and a simple search of patents in Google with the terms 'protein interaction network Alzheimer disease' retrieves more than 700 applications based on this approach. Network studies highlight the idea that multiple processes are involved in AD disease pathways, reveal new targets for further investigation and have generated new hypotheses. Network models have the potential to avoid biases inherent in other approaches, such as the emphasis on Aβ in animal models based on the more reductionist amyloid cascade hypothesis, and allow multiple disease pathways to be represented in a single model, perhaps better relating to the processes involved in human disease.

We have suggested that APP and its proteolytic fragments appear to dynamically modulate each other by multiple mechanisms with evidence for both direct and indirect feedback loops between Aβ, sAPPα, sAPPβ and full length APP. Additionally, the fragments derived from γ-cleavage, including the variable length fragments, P3 and Aβ, have the potential to interact with each other and modulate multiple neuronal processes in subtle and dynamic ways. Unfortunately, evidence for the role(s) of P3 is almost entirely missing from the literature and that for sAPPβ is limited. Without this information, we cannot have a full picture of the contributions of the APP proteolytic system to normal neuronal processes and how these change during disease progression.

Given the number and range of reciprocal feedback relationships that APP and its proteolytic fragments are involved in, the effects of perturbing the APP proteolytic system may be better understood as dynamic, synergistic actions involving multiple players. A wide range of neuronal processes have the potential to interact with and regulate APP processing, allowing APP to monitor the current state of the cell. The multiple proteolytic pathways allow APP to assimilate signals from wide-ranging cellular systems and pass these signals on via the ratios of APP fragments. The constant turnover of APP contributes to the ability of neurons to sense the current state of the cell and use this information to set in motion future responses. The potential for subtly different signals both from the α- and β-pathways via alternative peptides and the complex feedback relationships between them described above fits well with the fine control required for integrating neuronal systems.

The predicted synergy between APP and all its proteolytic fragments implies that the ratios of full length APP and all the various fragments, not just Aβ(1-40) and Aβ(1-42), are more important than absolute levels in coordinating neuronal responses. The complexity of the relationships described here has the potential to confound purely empirical investigations into the activities of any one proteolytic fragment studied in isolation. To avoid these predicted confounding effects, experimental approaches should be extended to account for the different concentrations, affinities and aggregation states of each peptide length, and experimental systems should be followed over time for each biological process in a systematic manner. With over 40 Aβ-type peptides [41], this presents a challenge.

Generating useful network models will require a more detailed and comprehensive approach to experimental design, involving a range of experimental controls to account for all the confounding factors. These data will be essential for generating network models that are truly relevant to our understanding of normal and abnormal processes in the context of the human population.

Abbreviations

Aβ:

amyloid beta protein

ACh:

acetylcholine

AChR:

acetylcholine receptor

AD:

Alzheimer's disease

APP:

amyloid precursor protein

BACE:

beta-site amyloid precursor protein cleaving enzyme

ECM:

extracellular matrix

IL:

interleukin

NF:

nuclear factor

PPI:

protein-protein interaction

PS:

presenilin

sAPP:

secreted amyloid precursor protein.

References

  1. Mirra SS, Heyman A, McKeel D, Sumi SM, Crain BJ, Brownlee LM, Vogel FS, Hughes JP, van Belle G, Berg L: The Consortium to Establish a Registry for Alzheimer's Disease (CERAD). Part II. Standardization of the neuropathologic assessment of Alzheimer's disease. Neurology. 1991, 41: 479-486.

    CAS  PubMed  Google Scholar 

  2. Braak H, Braak E: Neuropathological stageing of Alzheimer-related changes. Acta Neuropathol. 1991, 82: 239-259. 10.1007/BF00308809.

    CAS  PubMed  Google Scholar 

  3. Braak H, Braak E: Diagnostic criteria for neuropathologic assessment of Alzheimer's disease. Neurobiol Aging. 1997, 18 (4 Suppl): S85-88.

    CAS  PubMed  Google Scholar 

  4. Rossor MN, Fox NC, Freeborough PA, Harvey RJ: Clinical features of sporadic and familial Alzheimer's disease. Neurodegeneration. 1996, 5: 393-397. 10.1006/neur.1996.0052.

    CAS  PubMed  Google Scholar 

  5. Selkoe DJ: Alzheimer's disease: genes, proteins, and therapy. Physiol Rev. 2001, 81: 741-766.

    CAS  PubMed  Google Scholar 

  6. Zekanowski C, Religa D, Graff C, Filipek S, Kuźnicki J: Genetic aspects of Alzheimer's disease. Acta Neurobiol Exp (Wars). 2004, 64: 19-31.

    Google Scholar 

  7. Neuropathology Group. Medical Research Council Cognitive Function and Aging Study: Pathological correlates of late-onset dementia in a multicentre, community-based population in England and Wales. Neuropathology Group of the Medical Research Council Cognitive Function and Ageing Study (MRC CFAS). Lancet. 2001, 357: 169-75.

    Google Scholar 

  8. Savva GM, Wharton SB, Ince PG, Forster G, Matthews FE, Brayne C, Medical Research Council Cognitive Function and Ageing Study: Age, neuropathology, and dementia. N Engl J Med. 2009, 360: 2302-2309. 10.1056/NEJMoa0806142.

    CAS  PubMed  Google Scholar 

  9. Jellinger KA, Attems J: Neuropathological evaluation of mixed dementia. J Neurol Sci. 2007, 257: 80-87. 10.1016/j.jns.2007.01.045.

    CAS  PubMed  Google Scholar 

  10. Hardy JA, Higgins GA: Alzheimer's disease: the amyloid cascade hypothesis. Science. 1992, 256: 184-185. 10.1126/science.1566067.

    CAS  PubMed  Google Scholar 

  11. Hardy J, Selkoe DJ: The amyloid hypothesis of Alzheimer's disease: progress and problems on the road to therapeutics. Science. 2002, 297: 353-356. 10.1126/science.1072994.

    CAS  PubMed  Google Scholar 

  12. Selkoe DJ: Alzheimer's disease results from the cerebral accumulation and cytotoxicity of amyloid beta-protein. J Alzheimers Dis. 2001, 3: 75-80.

    CAS  PubMed  Google Scholar 

  13. Shen J, Kelleher RJ: The presenilin hypothesis of Alzheimer's disease: evidence for a loss-of-function pathogenic mechanism. Proc Natl Acad Sci USA. 2007, 104: 403-409. 10.1073/pnas.0608332104.

    PubMed Central  CAS  PubMed  Google Scholar 

  14. Koudinov AR, Koudinova NV: Cholesterol homeostasis failure as a unifying cause of synaptic degeneration. J Neurol Sci. 2005, 229-230: 233-240.

    CAS  PubMed  Google Scholar 

  15. Eckert A, Keil U, Marques CA, Bonert A, Frey C, Schüssel K, Müller WE: Mitochondrial dysfunction, apoptotic cell death, and Alzheimer's disease. Biochem Pharmacol. 2003, 66: 1627-1634. 10.1016/S0006-2952(03)00534-3.

    CAS  PubMed  Google Scholar 

  16. Hoyer S: The aging brain. Changes in the neuronal insulin/insulin receptor signal transduction cascade trigger late-onset sporadic Alzheimer disease (SAD). A mini-review. J Neural Transm. 2002, 109: 991-1002. 10.1007/s007020200082.

    CAS  PubMed  Google Scholar 

  17. Hoyer S: Causes and consequences of disturbances of cerebral glucose metabolism in sporadic Alzheimer disease: therapeutic implications. Adv Exp Med Biol. 2004, 541: 135-152. 10.1007/978-1-4419-8969-7_8.

    CAS  PubMed  Google Scholar 

  18. Bartus RT, Dean RL, Beer B, Lippa AS: The cholinergic hypothesis of geriatric memory dysfunction. Science. 1982, 217: 408-414. 10.1126/science.7046051.

    CAS  PubMed  Google Scholar 

  19. Mattson MP: Calcium and neurodegeneration. Aging Cell. 2007, 6: 337-350. 10.1111/j.1474-9726.2007.00275.x.

    CAS  PubMed  Google Scholar 

  20. Bojarski L, Herms J, Kuznicki J: Calcium dysregulation in Alzheimer's disease. Neurochem Int. 2008, 52: 621-633. 10.1016/j.neuint.2007.10.002.

    CAS  PubMed  Google Scholar 

  21. Arendt T: Synaptic plasticity and cell cycle activation in neurons are alternative effector pathways: the 'Dr. Jekyll and Mr. Hyde concept' of Alzheimer's disease or the yin and yang of neuroplasticity. Prog Neurobiol. 2003, 71: 83-248. 10.1016/j.pneurobio.2003.09.007.

    PubMed  Google Scholar 

  22. Fotuhi M, Hachinski V, Whitehouse PJ: Changing perspectives regarding late-life dementia. Nat Rev Neurol. 2009, 5: 649-658. 10.1038/nrneurol.2009.175.

    PubMed  Google Scholar 

  23. Brayne C: Research and Alzheimer's disease: an epidemiological perspective. Psychol Med. 1993, 23: 287-296. 10.1017/S003329170002835X.

    CAS  PubMed  Google Scholar 

  24. Yuraszeck TM, Neveu P, Rodriguez-Fernandez M, Robinson A, Kosik KS, Doyle FJ: Vulnerabilities in the tau network and the role of ultrasensitive points in tau pathophysiology. PLoS Comput Biol. 2010, 6: e1000997-10.1371/journal.pcbi.1000997.

    PubMed Central  PubMed  Google Scholar 

  25. Perreau VM, Orchard S, Adlard PA, Bellingham SA, Cappai R, Ciccotosto GD, Cowie TF, Crouch PJ, Duce JA, Evin G, Faux NG, Hill AF, Hung YH, James SA, Li QX, Mok SS, Tew DJ, White AR, Bush AI, Hermjakob H, Masters CL: A domain level interaction network of amyloid precursor protein and Abeta of Alzheimer's disease. Proteomics. 2010, 10: 2377-2395. 10.1002/pmic.200900773.

    CAS  PubMed  Google Scholar 

  26. Mewes HW, Wachinger B, Stumpflen V: Perspectives of a systems biology of the synapse: how to transform an indefinite data space into a model?. Pharmacopsychiatry. 2010, 43 (Suppl 1): S2-8.

    PubMed  Google Scholar 

  27. Liu ZP, Wang Y, Zhang XS, Chen L: Identifying dysfunctional crosstalk of pathways in various regions of Alzheimer's disease brains. BMC Syst Biol. 2010, 4 (Suppl 2): S11-10.1186/1752-0509-4-S2-S11.

    PubMed Central  PubMed  Google Scholar 

  28. Kell DB: Towards a unifying, systems biology understanding of large-scale cellular death and destruction caused by poorly liganded iron: Parkinson's, Huntington's, Alzheimer's, prions, bactericides, chemical toxicology and others as examples. Arch Toxicol. 2010, 84: 825-889. 10.1007/s00204-010-0577-x.

    PubMed Central  CAS  PubMed  Google Scholar 

  29. Noorbakhsh F, Overall CM, Power C: Deciphering complex mechanisms in neurodegenerative diseases: the advent of systems biology. Trends Neurosci. 2009, 32: 88-100. 10.1016/j.tins.2008.10.003.

    CAS  PubMed  Google Scholar 

  30. Miller JA, Oldham MC, Geschwind DH: A systems level analysis of transcriptional changes in Alzheimer's disease and normal aging. J Neurosci. 2008, 28: 1410-1420. 10.1523/JNEUROSCI.4098-07.2008.

    PubMed Central  CAS  PubMed  Google Scholar 

  31. Ray M, Ruan J, Zhang W: Variations in the transcriptome of Alzheimer's disease reveal molecular networks involved in cardiovascular diseases. Genome Biol. 2008, 9: R148-10.1186/gb-2008-9-10-r148.

    PubMed Central  PubMed  Google Scholar 

  32. Miller JA, Horvath S, Geschwind DH: Divergence of human and mouse brain transcriptome highlights Alzheimer disease pathways. Proc Natl Acad Sci USA. 2010, 107: 12698-12703. 10.1073/pnas.0914257107.

    PubMed Central  CAS  PubMed  Google Scholar 

  33. Soler-López M, Zanzoni A, Lluís R, Stelzl U, Aloy P: Interactome mapping suggests new mechanistic details underlying Alzheimer's disease. Genome Res. 2011, 21: 364-376. 10.1101/gr.114280.110.

    PubMed Central  PubMed  Google Scholar 

  34. UNIPROTKB. [http://www.uniprot.org/]

  35. NCBI Entrez Gene. [http://www.ncbi.nlm.nih.gov/gene]

  36. IntAct. [http://ebi.ac.uk/intact/site/]

  37. Pubmed. [http://www.ncbi.nlm.nih.gov/pubmed/]

  38. Compendium. [http://compendium.open.ac.uk/]

  39. 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. 10.1002/neu.10203.

    CAS  PubMed  Google Scholar 

  40. Tozaki H, Matsumoto A, Kanno T, Nagai K, Nagata T, Yamamoto S, Nishizaki T: The inhibitory and facilitatory actions of amyloid-beta peptides on nicotinic ACh receptors and AMPA receptors. Biochem Biophys Res Commun. 2002, 294: 42-45. 10.1016/S0006-291X(02)00429-1.

    CAS  PubMed  Google Scholar 

  41. Wang R, Sweeney D, Gandy SE, Sisodia SS: The profile of soluble amyloid beta protein in cultured cell media. Detection and quantification of amyloid beta protein and variants by immunoprecipitation-mass spectrometry. J Biol Chem. 1996, 271: 31894-31902. 10.1074/jbc.271.50.31894.

    CAS  PubMed  Google Scholar 

  42. Bush AI, Pettingell WH, de Paradis M, Tanzi RE, Wasco W: The amyloid beta-protein precursor and its mammalian homologues. Evidence for a zincmodulated heparin-binding superfamily. J Biol Chem. 1994, 269: 26618-26621.

    CAS  PubMed  Google Scholar 

  43. Turner PR, O'Connor K, Tate WP, Abraham WC: Roles of amyloid precursor protein and its fragments in regulating neural activity, plasticity and memory. Prog Neurobiol. 2003, 70: 1-32. 10.1016/S0301-0082(03)00089-3.

    CAS  PubMed  Google Scholar 

  44. Perez RG, Soriano S, Hayes JD, Ostaszewski B, Xia W, Selkoe DJ, Chen X, Stokin GB, Koo EH: Mutagenesis identifies new signals for beta-amyloid precursor protein endocytosis, turnover, and the generation of secreted fragments, including Abeta42. J Biol Chem. 1999, 274: 18851-18856. 10.1074/jbc.274.27.18851.

    CAS  PubMed  Google Scholar 

  45. Pietrzik CU, Busse T, Merriam DE, Weggen S, Koo EH: The cytoplasmic domain of the LDL receptor-related protein regulates multiple steps in APP processing. EMBO J. 2002, 21: 5691-5700. 10.1093/emboj/cdf568.

    PubMed Central  CAS  PubMed  Google Scholar 

  46. Koo EH, Squazzo SL, Selkoe DJ, Koo CH: Trafficking of cell-surface amyloid beta-protein precursor. I. Secretion, endocytosis and recycling as detected by labeled monoclonal antibody. J Cell Sci. 1996, 109: 991-998.

    CAS  PubMed  Google Scholar 

  47. Savage MJ, Trusko SP, Howland DS, Pinsker LR, Mistretta S, Reaume AG, Greenberg BD, Siman R, Scott RW: Turnover of amyloid beta-protein in mouse brain and acute reduction of its level by phorbol ester. J Neurosci. 1998, 18: 1743-1752.

    CAS  PubMed  Google Scholar 

  48. Lyckman AW, Confaloni AM, Thinakaran G, Sisodia SS, Moya KL: Post-translational processing and turnover kinetics of presynaptically targeted amyloid precursor superfamily proteins in the central nervous system. J Biol Chem. 1998, 273: 11100-11106. 10.1074/jbc.273.18.11100.

    CAS  PubMed  Google Scholar 

  49. Morales-Corraliza J, Mazzella MJ, Berger JD, Diaz NS, Choi JH, Levy E, Matsuoka Y, Planel E, Mathews PM: In vivo turnover of tau and APP metabolites in the brains of wild-type and Tg2576 mice: greater stability of sAPP in the beta-amyloid depositing mice. PLoS ONE. 2009, 4: e7134-10.1371/journal.pone.0007134.

    PubMed Central  PubMed  Google Scholar 

  50. Selkoe DJ: Cell biology of the amyloid beta-protein precursor and the mechanism of Alzheimer's disease. Annu Rev Cell Biol. 1994, 10: 373-403. 10.1146/annurev.cb.10.110194.002105.

    CAS  PubMed  Google Scholar 

  51. Zhang YW, Thompson R, Zhang H, Xu H: APP processing in Alzheimer's disease. Mol Brain. 2011, 4: 3-10.1186/1756-6606-4-3.

    PubMed Central  CAS  PubMed  Google Scholar 

  52. Sun X, He G, Song W: BACE2, as a novel APP theta-secretase, is not responsible for the pathogenesis of Alzheimer's disease in Down syndrome. FASEB J. 2006, 20: 1369-1376. 10.1096/fj.05-5632com.

    CAS  PubMed  Google Scholar 

  53. Beher D, Hesse L, Masters CL, Multhaup G: Regulation of amyloid protein precursor (APP) binding to collagen and mapping of the binding sites on APP and collagen type I. J Biol Chem. 1996, 271: 1613-1620. 10.1074/jbc.271.3.1613.

    CAS  PubMed  Google Scholar 

  54. Breen KC: APP-collagen interaction is mediated by a heparin bridge mechanism. Mol Chem Neuropathol. 1992, 16: 109-121. 10.1007/BF03159964.

    CAS  PubMed  Google Scholar 

  55. Hoe HS, Tran TS, Matsuoka Y, Howell BW, Rebeck GW: DAB1 and Reelin effects on amyloid precursor protein and ApoE receptor 2 trafficking and processing. J Biol Chem. 2006, 281: 35176-35185. 10.1074/jbc.M602162200.

    CAS  PubMed  Google Scholar 

  56. Hoe HS, Lee KJ, Carney RS, Lee J, Markova A, Lee JY, Howell BW, Hyman BT, Pak DT, Bu G, Rebeck GW: Interaction of reelin with amyloid precursor protein promotes neurite outgrowth. J Neurosci. 2009, 29: 7459-7473. 10.1523/JNEUROSCI.4872-08.2009.

    PubMed Central  CAS  PubMed  Google Scholar 

  57. Gralle M, Ferreira ST: Structure and functions of the human amyloid precursor protein: the whole is more than the sum of its parts. Prog Neurobiol. 2007, 82: 11-32. 10.1016/j.pneurobio.2007.02.001.

    CAS  PubMed  Google Scholar 

  58. Chautard E, Fatoux-Ardore M, Ballut L, Thierry-Mieg N, Ricard-Blum S: MatrixDB, the extracellular matrix interaction database. Nucleic Acids Res. 2011, 39 (Database issue): D235-240.

    PubMed Central  CAS  PubMed  Google Scholar 

  59. Dahms SO, Hoefgen S, Roeser D, Schlott B, Gührs KH, Than ME: Structure and biochemical analysis of the heparin-induced E1 dimer of the amyloid precursor protein. Proc Natl Acad Sci USA. 2010, 107: 5381-5386. 10.1073/pnas.0911326107.

    PubMed Central  CAS  PubMed  Google Scholar 

  60. Gralle M, Oliveira CL, Guerreiro LH, McKinstry WJ, Galatis D, Masters CL, Cappai R, Parker MW, Ramos CH, Torriani I, Ferreira ST: Solution conformation and heparin-induced dimerization of the full-length extracellular domain of the human amyloid precursor protein. J Mol Biol. 2006, 357: 493-508. 10.1016/j.jmb.2005.12.053.

    CAS  PubMed  Google Scholar 

  61. Multhaup G: Amyloid precursor protein and BACE function as oligomers. Neurodegener Dis. 2006, 3: 270-274. 10.1159/000095266.

    CAS  PubMed  Google Scholar 

  62. Gralle M, Botelho MG, Wouters FS: Neuroprotective secreted amyloid precursor protein acts by disrupting amyloid precursor protein dimers. J Biol Chem. 2009, 284: 15016-15025. 10.1074/jbc.M808755200.

    PubMed Central  CAS  PubMed  Google Scholar 

  63. Furukawa K, Sopher BL, Rydel RE, Begley JG, Pham DG, Martin GM, Fox M, Mattson MP: Increased activity-regulating and neuroprotective efficacy of alpha-secretase-derived secreted amyloid precursor protein conferred by a C-terminal heparin-binding domain. J Neurochem. 1996, 67: 1882-1896.

    CAS  PubMed  Google Scholar 

  64. Copanaki E, Chang S, Vlachos A, Tschäpe JA, Müller UC, Kögel D, Deller T: sAPPalpha antagonizes dendritic degeneration and neuron death triggered by proteasomal stress. Mol Cell Neurosci. 2010, 44: 386-393. 10.1016/j.mcn.2010.04.007.

    CAS  PubMed  Google Scholar 

  65. Sennvik K, Fastbom J, Blomberg M, Wahlund LO, Winblad B, Benedikz E: Levels of alpha- and beta-secretase cleaved amyloid precursor protein in the cerebrospinal fluid of Alzheimer's disease patients. Neurosci Lett. 2000, 278: 169-172. 10.1016/S0304-3940(99)00929-5.

    CAS  PubMed  Google Scholar 

  66. Bame KJ, Danda J, Hassall A, Tumova S: Abeta(1-40) prevents heparanasecatalyzed degradation of heparan sulfate glycosaminoglycans and proteoglycans in vitro. A role for heparan sulfate proteoglycan turnover in Alzheimer's disease. J Biol Chem. 1997, 272: 17005-17011. 10.1074/jbc.272.27.17005.

    CAS  PubMed  Google Scholar 

  67. Klajnert B, Cortijo-Arellano M, Bryszewska M, Cladera J: Influence of heparin and dendrimers on the aggregation of two amyloid peptides related to Alzheimer's and prion diseases. Biochem Biophys Res Commun. 2006, 339: 577-582. 10.1016/j.bbrc.2005.11.053.

    CAS  PubMed  Google Scholar 

  68. Bergamaschini L, Donarini C, Rossi E, De Luigi A, Vergani C, De Simoni MG: Heparin attenuates cytotoxic and inflammatory activity of Alzheimer amyloid-beta in vitro. Neurobiol Aging. 2002, 23: 531-536. 10.1016/S0197-4580(02)00003-9.

    CAS  PubMed  Google Scholar 

  69. Winkler K, Scharnagl H, Tisljar U, Hoschützky H, Friedrich I, Hoffmann MM, Hüttinger M, Wieland H, März W: Competition of Abeta amyloid peptide and apolipoprotein E for receptor-mediated endocytosis. J Lipid Res. 1999, 40: 447-455.

    CAS  PubMed  Google Scholar 

  70. Beckman M, Holsinger RM, Small DH: Heparin activates beta-secretase (BACE1) of Alzheimer's disease and increases autocatalysis of the enzyme. Biochemistry. 2006, 45: 6703-6714. 10.1021/bi052498t.

    CAS  PubMed  Google Scholar 

  71. White AR, Maher F, Brazier MW, Jobling MF, Thyer J, Stewart LR, Thompson A, Gibson R, Masters CL, Multhaup G, Beyreuther K, Barrow CJ, Collins SJ, Cappai R: Diverse fibrillar peptides directly bind the Alzheimer's amyloid precursor protein and amyloid precursor-like protein 2 resulting in cellular accumulation. Brain Res. 2003, 966: 231-244. 10.1016/S0006-8993(02)04173-2.

    CAS  PubMed  Google Scholar 

  72. Talamagas AA, Efthimiopoulos S, Tsilibary EC, Figueiredo-Pereira ME, Tzinia AK: Abeta(1-40)-induced secretion of matrix metalloproteinase-9 results in sAPPalpha release by association with cell surface APP. Neurobiol Dis. 2007, 28: 304-315. 10.1016/j.nbd.2007.07.016.

    CAS  PubMed  Google Scholar 

  73. Vitolo OV, Sant'Angelo A, Costanzo V, Battaglia F, Arancio O, Shelanski M: Amyloid beta -peptide inhibition of the PKA/CREB pathway and long-term potentiation: reversibility by drugs that enhance cAMP signaling. Proc Natl Acad Sci USA. 2002, 99: 13217-13221. 10.1073/pnas.172504199.

    PubMed Central  CAS  PubMed  Google Scholar 

  74. Ling X, Martins RN, Racchi M, Craft S, Helmerhorst E: Amyloid beta antagonizes insulin promoted secretion of the amyloid beta protein precursor. J Alzheimers Dis. 2002, 4: 369-374.

    CAS  PubMed  Google Scholar 

  75. Canet-Aviles RM, Anderton M, Hooper NM, Turner AJ, Vaughan PF: Muscarine enhances soluble amyloid precursor protein secretion in human neuroblastoma SH-SY5Y by a pathway dependent on protein kinase C(alpha), src-tyrosine kinase and extracellular signal-regulated kinase but not phospholipase C. Brain Res Mol Brain Res. 2002, 102: 62-72.

    CAS  PubMed  Google Scholar 

  76. Henriques AG, Vieira SI, da Cruz e Silva EF, da Cruz e Silva OA: Abeta hinders nuclear targeting of AICD and Fe65 in primary neuronal cultures. J Mol Neurosci. 2009, 39: 248-255. 10.1007/s12031-009-9192-9.

    PubMed Central  CAS  PubMed  Google Scholar 

  77. Barger SW, Mattson MP: Participation of gene expression in the protection against amyloid beta-peptide toxicity by the beta-amyloid precursor protein. Ann N Y Acad Sci. 1996, 777: 303-309. 10.1111/j.1749-6632.1996.tb34437.x.

    CAS  PubMed  Google Scholar 

  78. Cirrito JR, Kang JE, Lee J, Stewart FR, Verges DK, Silverio LM, Bu G, Mennerick S, Holtzman DM: Endocytosis is required for synaptic activity-dependent release of amyloid-beta in vivo. Neuron. 2008, 58: 42-51. 10.1016/j.neuron.2008.02.003.

    PubMed Central  CAS  PubMed  Google Scholar 

  79. Schöbel S, Neumann S, Hertweck M, Dislich B, Kuhn PH, Kremmer E, Seed B, Baumeister R, Haass C, Lichtenthaler SF: A novel sorting nexin modulates endocytic trafficking and alpha-secretase cleavage of the amyloid precursor protein. J Biol Chem. 2008, 283: 14257-14268. 10.1074/jbc.M801531200.

    PubMed  Google Scholar 

  80. Thal DR, Sassin I, Schultz C, Haass C, Braak E, Braak H: Fleecy amyloid deposits in the internal layers of the human entorhinal cortex are comprised of N-terminal truncated fragments of Abeta. J Neuropathol Exp Neurol. 1999, 58: 210-216. 10.1097/00005072-199902000-00010.

    CAS  PubMed  Google Scholar 

  81. Miravalle L, Calero M, Takao M, Roher AE, Ghetti B, Vidal R: Amino-terminally truncated Abeta peptide species are the main component of cotton wool plaques. Biochemistry. 2005, 44: 10810-10821. 10.1021/bi0508237.

    CAS  PubMed  Google Scholar 

  82. Zhao JH, Liu HL, Liu YF, Lin HY, Fang HW, Ho Y, Tsai WB: Molecular dynamics simulations to investigate the aggregation behaviors of the Abeta(17-42) oligomers. J Biomol Struct Dyn. 2009, 26: 481-490.

    CAS  PubMed  Google Scholar 

  83. Cordy JM, Hooper NM, Turner AJ: The involvement of lipid rafts in Alzheimer's disease. Mol Membr Biol. 2006, 23: 111-122. 10.1080/09687860500496417.

    CAS  PubMed  Google Scholar 

  84. Kamenetz F, Tomita T, Hsieh H, Seabrook G, Borchelt D, Iwatsubo T, Sisodia S, Malinow R: APP processing and synaptic function. Neuron. 2003, 37: 925-937. 10.1016/S0896-6273(03)00124-7.

    CAS  PubMed  Google Scholar 

  85. Pierrot N, Ghisdal P, Caumont AS, Octave JN: Intraneuronal amyloid-beta1-42 production triggered by sustained increase of cytosolic calcium concentration induces neuronal death. J Neurochem. 2004, 88: 1140-1150. 10.1046/j.1471-4159.2003.02227.x.

    CAS  PubMed  Google Scholar 

  86. Zhang X, Zhou K, Wang R, Cui J, Lipton SA, Liao FF, Xu H, Zhang YW: Hypoxia-inducible factor 1alpha (HIF-1alpha)-mediated hypoxia increases BACE1 expression and beta-amyloid generation. J Biol Chem. 2007, 282: 10873-10880. 10.1074/jbc.M608856200.

    CAS  PubMed  Google Scholar 

  87. Cantara S, Donnini S, Morbidelli L, Giachetti A, Schulz R, Memo M, Ziche M: Physiological levels of amyloid peptides stimulate the angiogenic response through FGF-2. FASEB J. 2004, 18: 1943-1945.

    CAS  PubMed  Google Scholar 

  88. Yan Y, Wang C: Abeta42 is more rigid than Abeta40 at the C terminus: implications for Abeta aggregation and toxicity. J Mol Biol. 2006, 364: 853-862. 10.1016/j.jmb.2006.09.046.

    CAS  PubMed  Google Scholar 

  89. Sgourakis NG, Yan Y, McCallum SA, Wang C, Garcia AE: The Alzheimer's peptides Abeta40 and 42 adopt distinct conformations in water: a combined MD/NMR study. J Mol Biol. 2007, 368: 1448-1457. 10.1016/j.jmb.2007.02.093.

    PubMed Central  CAS  PubMed  Google Scholar 

  90. Zou K, Kim D, Kakio A, Byun K, Gong JS, Kim J, Kim M, Sawamura N, Nishimoto S, Matsuzaki K, Lee B, Yanagisawa K, Michikawa M: Amyloid beta-protein (Abeta)1-40 protects neurons from damage induced by Abeta1-42 in culture and in rat brain. J Neurochem. 2003, 87: 609-619. 10.1046/j.1471-4159.2003.02018.x.

    CAS  PubMed  Google Scholar 

  91. Yamamoto N, Matsubara E, Maeda S, Minagawa H, Takashima A, Maruyama W, Michikawa M, Yanagisawa K: A ganglioside-induced toxic soluble Abeta assembly. Its enhanced formation from Abeta bearing the Arctic mutation. J Biol Chem. 2007, 282: 2646-2655.

    CAS  PubMed  Google Scholar 

  92. Heo C, Chang KA, Choi HS, Kim HS, Kim S, Liew H, Kim JA, Yu E, Ma J, Suh YH: Effects of the monomeric, oligomeric, and fibrillar Abeta42 peptides on the proliferation and differentiation of adult neural stem cells from subventricular zone. J Neurochem. 2007, 102: 493-500. 10.1111/j.1471-4159.2007.04499.x.

    CAS  PubMed  Google Scholar 

  93. Michikawa M, Gong JS, Fan QW, Sawamura N, Yanagisawa K: A novel action of alzheimer's amyloid beta-protein (Abeta): oligomeric Abeta promotes lipid release. J Neurosci. 2001, 21: 7226-7235.

    CAS  PubMed  Google Scholar 

  94. Schmid AW, Freir DB, Herron CE: Inhibition of LTP in vivo by beta-amyloid peptide in different conformational states. Brain Res. 2008, 1197: 135-142.

    CAS  PubMed  Google Scholar 

  95. Wang HY, Lee DH, Davis CB, Shank RP: Amyloid peptide Abeta(1-42) binds selectively and with picomolar affinity to alpha7 nicotinic acetylcholine receptors. J Neurochem. 2000, 75: 1155-1161.

    CAS  PubMed  Google Scholar 

  96. Alberghina L, Colangelo AM: The modular systems biology approach to investigate the control of apoptosis in Alzheimer's disease neurodegeneration. BMC Neurosci. 2006, 7 (Suppl 1): S2-10.1186/1471-2202-7-S1-S2.

    PubMed Central  PubMed  Google Scholar 

  97. Simpson JE, Ince PG, Haynes LJ, Theaker R, Gelsthorpe C, Baxter L, Forster G, Lace GL, Shaw PJ, Matthews FE, Savva GM, Brayne C, Wharton SB, MRC Cognitive Function and Ageing Neuropathology Study Group: Population variation in oxidative stress and astrocyte DNA damage in relation to Alzheimer-type pathology in the ageing brain. Neuropathol Appl Neurobiol. 2010, 36: 25-40. 10.1111/j.1365-2990.2009.01030.x.

    CAS  PubMed  Google Scholar 

  98. Simpson JE, Ince PG, Lace G, Forster G, Shaw PJ, Matthews F, Savva G, Brayne C, Wharton SB, MRC Cognitive Function and Ageing Neuropathology Study Group: Astrocyte phenotype in relation to Alzheimer-type pathology in the ageing brain. Neurobiol Aging. 2010, 31: 578-590. 10.1016/j.neurobiolaging.2008.05.015.

    CAS  PubMed  Google Scholar 

  99. Abbott JJ, Howlett DR, Francis PT, Williams RJ: Abeta(1-42) modulation of Akt phosphorylation via alpha7 nAChR and NMDA receptors. Neurobiol Aging. 2008, 29: 992-1001. 10.1016/j.neurobiolaging.2007.01.003.

    CAS  PubMed  Google Scholar 

  100. 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: 10639-10649. 10.1074/jbc.M806508200.

    PubMed Central  CAS  PubMed  Google Scholar 

  101. Zhao D, Watson JB, Xie CW: Amyloid beta prevents activation of calcium/calmodulin-dependent protein kinase II and AMPA receptor phosphorylation during hippocampal long-term potentiation. J Neurophysiol. 2004, 92: 2853-2858. 10.1152/jn.00485.2004.

    CAS  PubMed  Google Scholar 

  102. Dodart JC, Bales KR, Johnstone EM, Little SP, Paul SM: Apolipoprotein E alters the processing of the beta-amyloid precursor protein in APP(V717F) transgenic mice. Brain Res. 2002, 955: 191-199. 10.1016/S0006-8993(02)03437-6.

    CAS  PubMed  Google Scholar 

  103. Zlokovic BV, Yamada S, Holtzman D, Ghiso J, Frangione B: Clearance of amyloid beta-peptide from brain: transport or metabolism?. Nat Med. 2000, 6: 718-719.

    CAS  Google Scholar 

  104. Holtzman DM, Fagan AM, Mackey B, Tenkova T, Sartorius L, Paul SM, Bales K, Ashe KH, Irizarry MC, Hyman BT: Apolipoprotein E facilitates neuritic and cerebrovascular plaque formation in an Alzheimer's disease model. Ann Neurol. 2000, 47: 739-747. 10.1002/1531-8249(200006)47:6<739::AID-ANA6>3.0.CO;2-8.

    CAS  PubMed  Google Scholar 

  105. Tamamizu-Kato S, Cohen JK, Drake CB, Kosaraju MG, Drury J, Narayanaswami V: Interaction with amyloid beta peptide compromises the lipid binding function of apolipoprotein E. Biochemistry. 2008, 47: 5225-5234. 10.1021/bi702097s.

    CAS  PubMed  Google Scholar 

  106. Furukawa K, Barger SW, Blalock EM, Mattson MP: Activation of K+ channels and suppression of neuronal activity by secreted beta-amyloid-precursor protein. Nature. 1996, 379: 74-78. 10.1038/379074a0.

    CAS  PubMed  Google Scholar 

  107. Matsuda S, Matsuda Y, D'Adamio L: CD74 interacts with APP and suppresses the production of Abeta. Mol Neurodegener. 2009, 4: 41-10.1186/1750-1326-4-41.

    PubMed Central  PubMed  Google Scholar 

  108. Beel AJ, Mobley CK, Kim HJ, Tian F, Hadziselimovic A, Jap B, Prestegard JH, Sanders CR: Structural studies of the transmembrane C-terminal domain of the amyloid precursor protein (APP): does APP function as a cholesterol sensor?. Biochemistry. 2008, 47: 9428-9446. 10.1021/bi800993c.

    PubMed Central  CAS  PubMed  Google Scholar 

  109. Liu WW, Todd S, Coulson DT, Irvine GB, Passmore AP, McGuinness B, McConville M, Craig D, Johnston JA: A novel reciprocal and biphasic relationship between membrane cholesterol and beta-secretase activity in SH-SY5Y cells and in human platelets. J Neurochem. 2009, 108: 341-349. 10.1111/j.1471-4159.2008.05753.x.

    CAS  PubMed  Google Scholar 

  110. Abad-Rodriguez J, Ledesma MD, Craessaerts K, Perga S, Medina M, Delacourte A, Dingwall C, De Strooper B, Dotti CG: Neuronal membrane cholesterol loss enhances amyloid peptide generation. J Cell Biol. 2004, 167: 953-960. 10.1083/jcb.200404149.

    PubMed Central  CAS  PubMed  Google Scholar 

  111. Avdulov NA, Chochina SV, Igbavboa U, Warden CS, Vassiliev AV, Wood WG: Lipid binding to amyloid beta-peptide aggregates: preferential binding of cholesterol as compared with phosphatidylcholine and fatty acids. J Neurochem. 1997, 69: 1746-1752.

    CAS  PubMed  Google Scholar 

  112. Kristofiková Z, Kopecký V, Hofbauerová K, Hovorková P, Rípová D: Complex of amyloid beta peptides with 24-hydroxycholesterol and its effect on hemicholinium-3 sensitive carriers. Neurochem Res. 2008, 33: 412-421. 10.1007/s11064-007-9443-5.

    PubMed  Google Scholar 

  113. Shen Y, Sullivan T, Lee CM, Meri S, Shiosaki K, Lin CW: Induced expression of neuronal membrane attack complex and cell death by Alzheimer's beta-amyloid peptide. Brain Res. 1998, 796: 187-197. 10.1016/S0006-8993(98)00346-1.

    CAS  PubMed  Google Scholar 

  114. Mudher A, Chapman S, Richardson J, Asuni A, Gibb G, Pollard C, Killick R, Iqbal T, Raymond L, Varndell I, Sheppard P, Makoff A, Gower E, Soden PE, Lewis P, Murphy M, Golde TE, Rupniak HT, Anderton BH, Lovestone S: Dishevelled regulates the metabolism of amyloid precursor protein via protein kinase C/mitogen-activated protein kinase and c-Jun terminal kinase. J Neurosci. 2001, 21: 4987-4995.

    CAS  PubMed  Google Scholar 

  115. Sánchez-Alavez M, Chan SL, Mattson MP, Criado JR: Electrophysiological and cerebrovascular effects of the alpha-secretase-derived form of amyloid precursor protein in young and middle-aged rats. Brain Res. 2007, 1131: 112-117.

    PubMed  Google Scholar 

  116. Sepulveda FJ, Opazo C, Aguayo LG: Alzheimer beta-amyloid blocks epileptiform activity in hippocampal neurons. Mol Cell Neurosci. 2009, 41: 420-428. 10.1016/j.mcn.2009.04.012.

    CAS  PubMed  Google Scholar 

  117. Sabo SL, Ikin AF, Buxbaum JD, Greengard P: The Alzheimer amyloid precursor protein (APP) and FE65, an APP-binding protein, regulate cell movement. J Cell Biol. 2001, 153: 1403-1414. 10.1083/jcb.153.7.1403.

    PubMed Central  CAS  PubMed  Google Scholar 

  118. Kimberly WT, Zheng JB, Guénette SY, Selkoe DJ: The intracellular domain of the beta-amyloid precursor protein is stabilized by Fe65 and translocates to the nucleus in a notch-like manner. J Biol Chem. 2001, 276: 40288-40292.

    CAS  PubMed  Google Scholar 

  119. Ando K, Iijima KI, Elliott JI, Kirino Y, Suzuki T: Phosphorylation-dependent regulation of the interaction of amyloid precursor protein with Fe65 affects the production of beta-amyloid. J Biol Chem. 2001, 276: 40353-40361.

    CAS  PubMed  Google Scholar 

  120. Santiard-Baron D, Langui D, Delehedde M, Delatour B, Schombert B, Touchet N, Tremp G, Paul MF, Blanchard V, Sergeant N, Delacourte A, Duyckaerts C, Pradier L, Mercken L: Expression of human FE65 in amyloid precursor protein transgenic mice is associated with a reduction in beta-amyloid load. J Neurochem. 2005, 93: 330-338. 10.1111/j.1471-4159.2005.03026.x.

    CAS  PubMed  Google Scholar 

  121. Zhou D, Zambrano N, Russo T, D'Adamio L: Phosphorylation of a tyrosine in the amyloid-beta protein precursor intracellular domain inhibits Fe65 binding and signaling. J Alzheimers Dis. 2009, 16: 301-307.

    CAS  PubMed  Google Scholar 

  122. Hwang EM, Kim SK, Sohn JH, Lee JY, Kim Y, Kim YS, Mook-Jung I: Furin is an endogenous regulator of alpha-secretase associated APP processing. Biochem Biophys Res Commun. 2006, 349: 654-659. 10.1016/j.bbrc.2006.08.077.

    CAS  PubMed  Google Scholar 

  123. Mattson MP, Guo ZH, Geiger JD: Secreted form of amyloid precursor protein enhances basal glucose and glutamate transport and protects against oxidative impairment of glucose and glutamate transport in synaptosomes by a cyclic GMP-mediated mechanism. J Neurochem. 1999, 73: 532-537.

    CAS  PubMed  Google Scholar 

  124. Furukawa K, Mattson MP: Secreted amyloid precursor protein alpha selectively suppresses N-methyl-D-aspartate currents in hippocampal neurons: involvement of cyclic GMP. Neuroscience. 1998, 83: 429-438.

    CAS  PubMed  Google Scholar 

  125. Taylor CJ, Ireland DR, Ballagh I, Bourne K, Marechal NM, Turner PR, Bilkey DK, Tate WP, Abraham WC: Endogenous secreted amyloid precursor protein-alpha regulates hippocampal NMDA receptor function, long-term potentiation and spatial memory. Neurobiol Dis. 2008, 31: 250-260. 10.1016/j.nbd.2008.04.011.

    CAS  PubMed  Google Scholar 

  126. Suo Z, Wu M, Citron BA, Wong GT, Festoff BW: Abnormality of G-protein-coupled receptor kinases at prodromal and early stages of Alzheimer's disease: an association with early beta-amyloid accumulation. J Neurosci. 2004, 24: 3444-3452. 10.1523/JNEUROSCI.4856-03.2004.

    CAS  PubMed  Google Scholar 

  127. Nishimoto I, Okamoto T, Matsuura Y, Takahashi S, Okamoto T, Murayama Y, Ogata E: Alzheimer amyloid protein precursor complexes with brain GTP-binding protein G(o). Nature. 1993, 362: 75-79. 10.1038/362075a0.

    CAS  PubMed  Google Scholar 

  128. Okamoto T, Takeda S, Murayama Y, Ogata E, Nishimoto I: Ligand-dependent G protein coupling function of amyloid transmembrane precursor. J Biol Chem. 1995, 270: 4205-4208. 10.1074/jbc.270.9.4205.

    CAS  PubMed  Google Scholar 

  129. Shaked GM, Chauv S, Ubhi K, Hansen LA, Masliah E: Interactions between the amyloid precursor protein C-terminal domain and G proteins mediate calcium dysregulation and amyloid beta toxicity in Alzheimer's disease. FEBS J. 2009, 276: 2736-2751. 10.1111/j.1742-4658.2009.06997.x.

    PubMed Central  CAS  PubMed  Google Scholar 

  130. Koudinov AR, Berezov TT, Kumar A, Koudinova NV: Alzheimer's amyloid beta interaction with normal human plasma high density lipoprotein: association with apolipoprotein and lipids. Clin Chim Acta. 1998, 270: 75-84. 10.1016/S0009-8981(97)00207-6.

    CAS  PubMed  Google Scholar 

  131. Patey SJ, Edwards EA, Yates EA, Turnbull JE: Heparin derivatives as inhibitors of BACE-1, the Alzheimer's beta-secretase, with reduced activity against factor Xa and other proteases. J Med Chem. 2006, 49: 6129-6132. 10.1021/jm051221o.

    CAS  PubMed  Google Scholar 

  132. Patey SJ, Edwards EA, Yates EA, Turnbull JE: Engineered heparins: novel beta-secretase inhibitors as potential Alzheimer's disease therapeutics. Neurodegener Dis. 2008, 5: 197-199. 10.1159/000113701.

    CAS  PubMed  Google Scholar 

  133. Kong Q, Peterson TS, Baker O, Stanley E, Camden J, Seye CI, Erb L, Simonyi A, Wood WG, Sun GY, Weisman GA: Interleukin-1beta enhances nucleotideinduced and alpha-secretase-dependent amyloid precursor protein processing in rat primary cortical neurons via up-regulation of the P2Y(2) receptor. J Neurochem. 2009, 109: 1300-1310. 10.1111/j.1471-4159.2009.06048.x.

    PubMed Central  CAS  PubMed  Google Scholar 

  134. Tachida Y, Nakagawa K, Saito T, Saido TC, Honda T, Saito Y, Murayama S, Endo T, Sakaguchi G, Kato A, Kitazume S, Hashimoto Y: Interleukin-1 beta up-regulates TACE to enhance alpha-cleavage of APP in neurons: resulting decrease in Abeta production. J Neurochem. 2008, 104: 1387-1393. 10.1111/j.1471-4159.2007.05127.x.

    CAS  PubMed  Google Scholar 

  135. Sheng JG, Ito K, Skinner RD, Mrak RE, Rovnaghi CR, Van Eldik LJ, Griffin WS: In vivo and in vitro evidence supporting a role for the inflammatory cytokine interleukin-1 as a driving force in Alzheimer pathogenesis. Neurobiol Aging. 1996, 17: 761-766. 10.1016/0197-4580(96)00104-2.

    PubMed Central  CAS  PubMed  Google Scholar 

  136. Pérez A, Morelli L, Cresto JC, Castaño EM: Degradation of soluble amyloid beta-peptides 1-40, 1-42, and the Dutch variant 1-40Q by insulin degrading enzyme from Alzheimer disease and control brains. Neurochem Res. 2000, 25: 247-255. 10.1023/A:1007527721160.

    PubMed  Google Scholar 

  137. Young-Pearse TL, Chen AC, Chang R, Marquez C, Selkoe DJ: Secreted APP regulates the function of full-length APP in neurite outgrowth through interaction with integrin beta1. Neural Dev. 2008, 3: 15-10.1186/1749-8104-3-15.

    PubMed Central  PubMed  Google Scholar 

  138. Caltagarone J, Jing Z, Bowser R: Focal adhesions regulate Abeta signaling and cell death in Alzheimer's disease. Biochim Biophys Acta. 2007, 1772: 438-445.

    PubMed Central  CAS  PubMed  Google Scholar 

  139. Wang Q, Klyubin I, Wright S, Griswold-Prenner I, Rowan MJ, Anwyl R: Alpha v integrins mediate beta-amyloid induced inhibition of long-term potentiation. Neurobiol Aging. 2008, 29: 1485-1493. 10.1016/j.neurobiolaging.2007.03.018.

    CAS  PubMed  Google Scholar 

  140. Terzi E, Holzemann G, Seelig J: Interaction of Alzheimer beta-amyloid peptide(1-40) with lipid membranes. Biochemistry. 1997, 36: 14845-14852. 10.1021/bi971843e.

    CAS  PubMed  Google Scholar 

  141. Hertel C, Terzi E, Hauser N, Jakob-Rotne R, Seelig J, Kemp JA: Inhibition of the electrostatic interaction between beta-amyloid peptide and membranes prevents beta-amyloid-induced toxicity. Proc Natl Acad Sci USA. 1997, 94: 9412-9416. 10.1073/pnas.94.17.9412.

    PubMed Central  CAS  PubMed  Google Scholar 

  142. Verdier Y, Zarandi M, Penke B: Amyloid beta-peptide interactions with neuronal and glial cell plasma membrane: binding sites and implications for Alzheimer's disease. J Pept Sci. 2004, 10: 229-248. 10.1002/psc.573.

    CAS  PubMed  Google Scholar 

  143. Cordy JM, Hussain I, Dingwall C, Hooper NM, Turner AJ: Exclusively targeting beta-secretase to lipid rafts by GPI-anchor addition up-regulates beta-site processing of the amyloid precursor protein. Proc Natl Acad Sci USA. 2003, 100: 11735-11740. 10.1073/pnas.1635130100.

    PubMed Central  CAS  PubMed  Google Scholar 

  144. Saavedra L, Mohamed A, Ma V, Kar S, de Chaves EP: Internalization of beta-amyloid peptide by primary neurons in the absence of apolipoprotein E. J Biol Chem. 2007, 282: 35722-35732. 10.1074/jbc.M701823200.

    CAS  PubMed  Google Scholar 

  145. Sawamura N, Ko M, Yu W, Zou K, Hanada K, Suzuki T, Gong JS, Yanagisawa K, Michikawa M: Modulation of amyloid precursor protein cleavage by cellular sphingolipids. J Biol Chem. 2004, 279: 11984-11991. 10.1074/jbc.M309832200.

    CAS  PubMed  Google Scholar 

  146. Wu MN, He YX, Guo F, Qi JS: Alpha4beta2 nicotinic acetylcholine receptors are required for the amyloid beta protein-induced suppression of long-term potentiation in rat hippocampal CA1 region in vivo. Brain Res Bull. 2008, 77: 84-90. 10.1016/j.brainresbull.2008.06.005.

    CAS  PubMed  Google Scholar 

  147. Yamin G: NMDA receptor-dependent signaling pathways that underlie amyloid beta-protein disruption of LTP in the hippocampus. J Neurosci Res. 2009, 87: 1729-1736. 10.1002/jnr.21998.

    CAS  PubMed  Google Scholar 

  148. Caccamo A, Oddo S, Billings LM, Green KN, Martinez-Coria H, Fisher A, LaFerla FM: M1 receptors play a central role in modulating AD-like pathology in transgenic mice. Neuron. 2006, 49: 671-682. 10.1016/j.neuron.2006.01.020.

    CAS  PubMed  Google Scholar 

  149. Fisher A: M1 muscarinic agonists target major hallmarks of Alzheimer's disease - the pivotal role of brain M1 receptors. Neurodegener Dis. 2008, 5: 237-240. 10.1159/000113712.

    CAS  PubMed  Google Scholar 

  150. Nitsch RM, Slack BE, Farber SA, Schulz JG, Deng M, Kim C, Borghesani PR, Korver W, Wurtman RJ, Growdon JH: Regulation of proteolytic processing of the amyloid beta-protein precursor of Alzheimer's disease in transfected cell lines and in brain slices. J Neural Transm Suppl. 1994, 44: 21-27.

    CAS  PubMed  Google Scholar 

  151. Farber SA, Nitsch RM, Schulz JG, Wurtman RJ: Regulated secretion of beta-amyloid precursor protein in rat brain. J Neurosci. 1995, 15: 7442-7451.

    CAS  PubMed  Google Scholar 

  152. Liskowsky W, Schliebs R: Muscarinic acetylcholine receptor inhibition in transgenic Alzheimer-like Tg2576 mice by scopolamine favours the amyloidogenic route of processing of amyloid precursor protein. Int J Dev Neurosci. 2006, 24: 149-156. 10.1016/j.ijdevneu.2005.11.010.

    CAS  PubMed  Google Scholar 

  153. Zuchner T, Perez-Polo JR, Schliebs R: Beta-secretase BACE1 is differentially controlled through muscarinic acetylcholine receptor signaling. J Neurosci Res. 2004, 77: 250-257. 10.1002/jnr.20152.

    PubMed  Google Scholar 

  154. Kim C, Jang CH, Bang JH, Jung MW, Joo I, Kim SU, Mook-Jung I: Amyloid precursor protein processing is separately regulated by protein kinase C and tyrosine kinase in human astrocytes. Neurosci Lett. 2002, 324: 185-188. 10.1016/S0304-3940(02)00217-3.

    CAS  PubMed  Google Scholar 

  155. Qiu WQ, Ferreira A, Miller C, Koo EH, Selkoe DJ: Cell-surface beta-amyloid precursor protein stimulates neurite outgrowth of hippocampal neurons in an isoform-dependent manner. J Neurosci. 1995, 15: 2157-2167.

    CAS  PubMed  Google Scholar 

  156. Rossner S, Sastre M, Bourne K, Lichtenthaler SF: Transcriptional and translational regulation of BACE1 expression - implications for Alzheimer's disease. Prog Neurobiol. 2006, 79: 95-111. 10.1016/j.pneurobio.2006.06.001.

    CAS  PubMed  Google Scholar 

  157. Choi S, Kim JH, Roh EJ, Ko MJ, Jung JE, Kim HJ: Nuclear factor-kappaB activated by capacitative Ca2+ entry enhances muscarinic receptor-mediated soluble amyloid precursor protein (sAPPalpha) release in SH-SY5Y cells. J Biol Chem. 2006, 281: 12722-12728. 10.1074/jbc.M601018200.

    CAS  PubMed  Google Scholar 

  158. Kihara T, Shimohama S, Urushitani M, Sawada H, Kimura J, Kume T, Maeda T, Akaike A: Stimulation of alpha4beta2 nicotinic acetylcholine receptors inhibits beta-amyloid toxicity. Brain Res. 1998, 792: 331-334. 10.1016/S0006-8993(98)00138-3.

    CAS  PubMed  Google Scholar 

  159. Nagele RG, D'Andrea MR, Anderson WJ, Wang HY: Intracellular accumulation of beta-amyloid(1-42) in neurons is facilitated by the alpha 7 nicotinic acetylcholine receptor in Alzheimer's disease. Neuroscience. 2002, 110: 199-211. 10.1016/S0306-4522(01)00460-2.

    CAS  PubMed  Google Scholar 

  160. Dougherty JJ, Wu J, Nichols RA: Beta-amyloid regulation of presynaptic nicotinic receptors in rat hippocampus and neocortex. J Neurosci. 2003, 23: 6740-6747.

    CAS  PubMed  Google Scholar 

  161. Wang HY, Lee DH, D'Andrea MR, Peterson PA, Shank RP, Reitz AB: beta- Amyloid(1-42) binds to alpha7 nicotinic acetylcholine receptor with high affinity. Implications for Alzheimer's disease pathology. J Biol Chem. 2000, 275: 5626-5632. 10.1074/jbc.275.8.5626.

    CAS  PubMed  Google Scholar 

  162. 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: 1051-1058. 10.1038/nn1503.

    CAS  PubMed  Google Scholar 

  163. Camden JM, Schrader AM, Camden RE, González FA, Erb L, Seye CI, Weisman GA: P2Y2 nucleotide receptors enhance alpha-secretase-dependent amyloid precursor protein processing. J Biol Chem. 2005, 280: 18696-18702. 10.1074/jbc.M500219200.

    CAS  PubMed  Google Scholar 

  164. Roncarati R, Sestan N, Scheinfeld MH, Berechid BE, Lopez PA, Meucci O, McGlade JC, Rakic P, D'Adamio L: The gamma-secretase-generated intracellular domain of beta-amyloid precursor protein binds Numb and inhibits Notch signaling. Proc Natl Acad Sci USA. 2002, 99: 7102-7107. 10.1073/pnas.102192599.

    PubMed Central  CAS  PubMed  Google Scholar 

  165. Etcheberrigaray R, Tan M, Dewachter I, Kuipéri C, Van der Auwera I, Wera S, Qiao L, Bank B, Nelson TJ, Kozikowski AP, Van Leuven F, Alkon DL: Therapeutic effects of PKC activators in Alzheimer's disease transgenic mice. Proc Natl Acad Sci USA. 2004, 101: 11141-11146. 10.1073/pnas.0403921101.

    PubMed Central  CAS  PubMed  Google Scholar 

  166. Rossner S, Mendla K, Schliebs R, Bigl V: Protein kinase Calpha and beta1 isoforms are regulators of alpha-secretory proteolytic processing of amyloid precursor protein in vivo. Eur J Neurosci. 2001, 13: 1644-1648. 10.1046/j.0953-816x.2001.01525.x.

    CAS  PubMed  Google Scholar 

  167. Favit A, Grimaldi M, Nelson TJ, Alkon DL: Alzheimer's-specific effects of soluble beta-amyloid on protein kinase C-alpha and -gamma degradation in human fibroblasts. Proc Natl Acad Sci USA. 1998, 95: 5562-5567. 10.1073/pnas.95.10.5562.

    PubMed Central  CAS  PubMed  Google Scholar 

  168. Durakoglugil MS, Chen Y, White CL, Kavalali ET, Herz J: Reelin signaling antagonizes beta-amyloid at the synapse. Proc Natl Acad Sci USA. 2009, 106: 15938-15943. 10.1073/pnas.0908176106.

    PubMed Central  CAS  PubMed  Google Scholar 

  169. Tamayev R, Zhou D, D'Adamio L: The interactome of the amyloid beta precursor protein family members is shaped by phosphorylation of their intracellular domains. Mol Neurodegener. 2009, 4: 28-10.1186/1750-1326-4-28.

    PubMed Central  PubMed  Google Scholar 

  170. Scheinfeld MH, Ghersi E, Davies P, D'Adamio L: Amyloid beta protein precursor is phosphorylated by JNK-1 independent of, yet facilitated by, JNK-interacting protein (JIP)-1. J Biol Chem. 2003, 278: 42058-42063. 10.1074/jbc.M304853200.

    CAS  PubMed  Google Scholar 

  171. Hoe HS, Cooper MJ, Burns MP, Lewis PA, van der Brug M, Chakraborty G, Cartagena CM, Pak DT, Cookson MR, Rebeck GW: The metalloprotease inhibitor TIMP-3 regulates amyloid precursor protein and apolipoprotein E receptor proteolysis. J Neurosci. 2007, 27: 10895-10905. 10.1523/JNEUROSCI.3135-07.2007.

    CAS  PubMed  Google Scholar 

  172. Stein TD, Johnson JA: Lack of neurodegeneration in transgenic mice overexpressing mutant amyloid precursor protein is associated with increased levels of transthyretin and the activation of cell survival pathways. J Neurosci. 2002, 22: 7380-7388.

    CAS  PubMed  Google Scholar 

  173. Costa R, Ferreira-da-Silva F, Saraiva MJ, Cardoso I: Transthyretin protects against A-beta peptide toxicity by proteolytic cleavage of the peptide: a mechanism sensitive to the Kunitz protease inhibitor. PLoS ONE. 2008, 3: e2899-10.1371/journal.pone.0002899.

    PubMed Central  PubMed  Google Scholar 

Download references

Acknowledgements

This work was supported by funding from the Cambridge and Peterborough CLAHRC.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Sally Hunter.

Additional information

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

SH wrote the paper in discussion and with contributions from CB.

Authors’ original submitted files for images

Below are the links to the authors’ original submitted files for images.

Authors’ original file for figure 1

Authors’ original file for figure 2

Rights and permissions

Reprints and permissions

About this article

Cite this article

Hunter, S., Brayne, C. Relationships between the amyloid precursor protein and its various proteolytic fragments and neuronal systems. Alz Res Therapy 4, 10 (2012). https://doi.org/10.1186/alzrt108

Download citation

  • Published:

  • DOI: https://doi.org/10.1186/alzrt108

Keywords