Abstract
Alzheimer’s disease (AD) is an age-related neurodegenerative disease that affects approximately 24 million people worldwide. A number of different risk factors have been implicated in AD; however, neuritic (amyloid) plaques are considered as one of the defining risk factors and pathological hallmarks of the disease. In the past decade, enormous efforts have been devoted to understand the genetics and molecular pathogenesis leading to neuronal death in AD, which has been transferred into extensive experimental approaches aimed at reversing disease progression. Modern medicine is facing an increasing number of treatments available for vascular and neurodegenerative brain diseases, but no causal or neuroprotective treatment has yet been established. Almost all neurological conditions are characterized by progressive neuronal dysfunction, which, regardless of the pathogenetic mechanism, finally leads to neuronal death. The particular emphasis of this review is on risk factors and mechanisms resulting in neuronal loss in AD and current and prospective opportunities for therapeutic interventions. This review discusses these issues with a view to inspiring the development of new agents that could be useful for the treatment of AD.
Keywords: Alzheimer’s disease, neuronal death, neurodegeneration, neurotoxicity, neuroprotection, amyloid plaques
Introduction
- Origin of the disease and mechanisms for neuronal death in AD
- Inflammatory reaction
- Aβ-induced neurotoxicity
- Complement-mediated neurodegeneration
- Oxidative and nitrosylative damage
- Proteasome inhibitor-induced neurotoxicity
- Cholesterol-induced neurotoxicity
- Viral infections
- Interrelationship between different aetiopathogenic theories of AD
- Treatment approaches
- Drugs which modify AD symptoms
- Cholinergic treatment
- Antiglutamic treatment
- β- and γ-secretase inhibitors
- Drugs which potentially prevent and modify AD symptoms
- Vitamins and antioxidants
- Non-steroidal anti-inflammatory drugs
- Oestrogen replacement therapy
- Inhibitors of complement-mediated degeneration
- Cobra venom factor (CVF)
- Heparin and other polyionic agents
- Other small molecule inhibitors
- Recombinant protein inhibitors
- Anti-amyloid therapy (‘vaccination’)
- Targeting proteasome inhibition
- Neuropeptide mixture (Cerebrolysin)
Conclusions
Introduction
Alzheimer disease (AD) is a neurodegenerative disease that causes changes in brain function. AD usually affects people over the age of 65 years, with a plethora of devastating clinical symptoms such as progressive decline in memory, thinking, language and learning capacity. Age is the strongest predictor for the development and progression of AD and with the rapidly aging population, AD clearly poses a major health problem and socio-economic burden. An estimated 5–10% of the population aged 65 years and over, and 40% of the population greater than 85 years of age are likely to be affected [1]. The genetic predisposition accounts for only 5–10% and is associated with early onset [2, 3].
Given the aging population in Western countries, the grave suffering of the many individuals concerned and their families, and the increasing economic cost of AD in terms of both healthcare expenses and lost wages it makes it important to understand the disease mechanisms and propose effective treatment strategies. The clinical manifestation of AD is considered to be correlated with the degree of neuronal loss in the brain and more specifically in hippocampal and neocortical areas. While AD can be identified based on clinical symptoms and by excluding other conditions with similar phenomenology and psychopathology, a definite diagnosis is only possible post-mortem by histological analysis of the patient’s brain. Extracellular senile (amyloid) plaques and intracellular neurofibrillary tangles (NFTs) are considered to be hallmarks of the disease [4]. Senile plaques contain extracellular deposits of amyloid-β protein (Aβ), α-synuclein, ubiquitin and apolipoprotein E. NFTs are intracellular and extracellular aggregates of hyperphosphorylated τ protein and apolipoprotein E [5]. Evidence that Aβ alone is not sufficient for initiating the pathophysiological cascade of AD came from experiments with transgenic mice, which suggested that sustained brain inflammation might be another essential factor in AD pathogenesis. Different inflammatory markers such as activated microglia and astrocytes, elevated levels of various cytokines and complement activation products are found in AD brains [6]. Complement proteins are reported to be integral components of amyloid plaques and cerebral vascular amyloid in AD. They are found at the earliest stages of amyloid deposition and their activation coincides with the progressive clinical manifestation of AD [7, 8].
Here we review the interrelationship between different risk factors for AD aetiology with particular emphasis on their role for neuronal death as well as different strategies for neuroprotection as a potentially promising therapeutic strategy.
Origin of the disease and mechanisms for neuronal death in AD
The pathogenesis of AD is not yet fully understood. Various hypotheses with respect to disease aetiology have been proposed and those that are most widely accepted on grounds of a solid experimental data base and clinical evidence are described below.
Inflammatory reaction
This hypothesis is based on observations that non-steroidal anti-inflammatory drugs (NSAIDs) can reduce neurotoxins in patients suffering from AD. In epidemiological and clinical studies, it was initially noted that patients on high doses of anti-inflammatory drugs exhibit a reduced incidence of AD and reduced the AD symptoms [9–14]. However, this hypothesis became even more attractive when it was demonstrated that the mediators and products of inflammatory reaction, such as cytokines [15, 16], complement proteins (reviewed in [17]), free radicals [18, 19], adhesion molecules [20, 21] and prostaglandins (reviewed in [22]), were neurotoxic in experiments with neural models. These products of inflammatory reactions may represent extracellular signals which initiate and promote neuronal degeneration in AD. For instance, long-time administration of NSAIDs to arthritic patients can reduce the risk of developing AD [7, 11]. Potent inflammatory molecules, such as cytokines, chemokines and complement factors, are present in both cerebrospinal fluid (CSF) and plaques from patients with AD [7]. Aggregated peptides, such as Aβ are shown to induce the production of pro-inflammatory cytokines like interleukin (IL)-1β, tumour necrosis factor (TNF)-α and chemokines (MIP-1α, MIP-1β, MCP-1) by microglial cells [23–25]. These inflammatory molecules were found to be toxic in neuronal models of AD and were implicated in extracellular signalling which initiated and promoted neuronal degeneration in AD. Several intracellular signals are candidates for mediating actions of inflammatory factors inside the cells such as Aβ[26, 27], ubiquitin [28, 29] and proteasome [30–32]. The observed protective effect of NSAIDs could be due to a reduction of microglial activation and/or decreased generation of Aβ[33, 34]. These observations are additionally confirmed by in vitro studies with human neuronal cells which seem to be protected from the toxic effects of Aβ by NSAIDs [35].
While epidemiological and experimental studies lend strong support for neuroinflammatory responses as drivers of AD pathogenesis, recent in vivo studies also support a beneficial role for such reactions (reviewed in [36]). A very strong support for the beneficial impact of neuroinflammation on neuronal survival and function came recently from a study with transgenic mice with brain-directed overexpression of human soluble IL-1 receptor antagonist [37]. Chronic blockade of IL-1 signalling in the brain of these animals was found associated with an atrophic phenotype of the brain and with modified levels of the amyloid precursor protein (APP) and presenilin 1 (PS1), a critical component of APP processing machinery (discussed below). A number of reports have provided evidence that activation of microglia and the subsequent degradation of amyloid plaques may underlie this phenomenon. These observations in animal models challenge earlier assumptions that IL-1 elevation and resulting neuroinflammatory processes play a purely detrimental role in AD, and prompt a need for new characterizations of IL-1 function.
Aβ-induced neurotoxicity
The extracellular Aβ deposition has attracted major attention as a cause of cytotoxicity in AD. The original ‘amyloid hypothesis’ argues that Aβ deposition is the initiator for AD pathogenesis, based on the following facts: Aβ is a major component of the amyloid plaques [38]; the deposition of Aβ occurs prior to other pathological events such as NFT formation and neuronal loss [39]; synthetic Aβ peptides, particularly Aβ1–42/43, induce neuronal death in vitro[40, 41]. Aβ is proteolytically derived from the β-APP by the action of β- and γ-secretases [42] (Fig. 1). These enzymes cleave at the N-terminus of the Aβ region of APP followed by cleavage at the C terminus producing peptides of a length of 39 to 43 amino acids. The two forms found predominantly in amyloid plaques and believed to be associated with the disease, are 40 and 42 amino acids long. Alternatively, the α-secretase cleaves APP within the Aβ domain yielding a non-toxic C terminal fragment, followed by γ-secretase cleavage resulting in a C terminal fragment-γ and a small peptide called P3 that is not considered amyloidogenic. Presenilins (PS1 and PS2) regulate the activity of γ-secretase and are involved in the processing of APP and in the generation of Aβ peptides [43] (Fig. 1). Presenilins were first identified as enzymes acting as γ-secretase [44]. More recently, it was discovered that presenilins are part of a tetrameric complex comprised the proteins nicastrin, Aph1 (anterior pharynx-defective phenotype) and Pen2 (presenilin enhancer) that altogether regulate the production of Aβ peptides [45, 46]. In AD patients carrying missense mutations on either of the genes for PS1 or PS2, it was reported that the overall levels of Aβ peptides were increased in the plasma, and that levels of the more amyloidogenic Aβ42 were increased in cultured fibroblasts of patients carrying inherited mutations on the genes for presenilins [47–49]. Experiments with animal models lacking expression of either PS1 or PS2 showed that between the two presenilins PS1 plays a major role regulating the activity of γ-secretase and thus Aβ formation [50, 51]. This new knowledge opens new hypotheses about the mechanism that orchestrates the regulated intra-membrane proteolysis in γ-secretase substrates and in APP in particular.
Figure 1.

Processing of APP. APP can be processed by different sets of enzymes: one pathway leads to amyloid plaque formation (amyloidogenic), while the other does not (non-amyloidogenic). In the non-amyloidogenic pathway, APP is cleaved first by α-secretase to yield a soluble N-terminal fragment and a C-terminal fragment integrated within the cellular membrane. The soluble protein may be involved in the enhancement of synaptogenesis, neurite outgrowth and neuronal survival, and is considered to be neuroprotective. The membrane fragment of the cleaved APP is acted upon by γ-secretase, a tetrameric complex comprised presenilins, nicastrin, Aph1 and Pen2, to yield a soluble N-terminal fragment (P3) and a membrane-bound C-terminal fragment. In the amyloidogenic pathway, APP is cleaved first by β-secretase, yielding a soluble N-terminal fragment and a membrane-bound C-terminal fragment. The membrane fragment is then cleaved by γ-secretase, yielding a membrane-bound C-terminal fragment and a soluble N-terminal fragment (Aβ). While Aβ is required for neuronal function, it can aggregate in the extracellular space of the brain to form amyloid plaques.
Interestingly, presenilins have been implicated not only in regulation of activity of γ-secretase but also in control of apoptotic machinery [52, 53]. AD associated presenilins showed to be substrates for proteolytic degradation. These investigations strongly suggest that alternative presenilin fragments could regulate cell survival. Furthermore, mutations in presenilin genes, which likely suppress proteolytic degradation of these proteins, sensitize neurons to apoptosis by different mechanisms (e.g. increased caspase-3 activation, production of oxyradicals, calcium signalling dysregulation). These data demonstrate the complex dual nature of regulation of neuronal death in AD by presenilins and suggest that any treatment targeting these proteins might be a double-edged sword and should be carefully considered.
Accumulated Aβ induces multiple cytotoxic effects, including oxidative stress, and alternation of ionic homeostasis in neurons [54, 55]. Aβ also alters the activities of various kinases, including GSK3β, cdk5, PKA and causes hyperphosphorylation of τ protein, leading to NFT formation [56–58]. These Aβ-initiated toxicities directly or indirectly induce neuronal cell death. Although this classical Aβ hypothesis does explain some of the mechanisms underlying the pathogenesis and progression of AD, there is also evidence against this hypothesis. For example, the quantity of Aβ deposits does not correlate with clinical features, as senile plaques are also found in brains of elderly subjects without dementia [59]. Accumulation of senile plaques does not necessarily correlate with the amount of synaptic loss [60, 61] and the severity of the clinical manifestation [62]. In addition, several lines of transgenic mice with human familial AD mutant genes show considerable Aβ deposits in brain without exhibiting other AD-specific pathological features or behavioural abnormalities. Even though some evidence suggest that the Aβ deposition alone is not sufficient for the development of AD, formation of the senile plaques seems to be involved in triggering most of the subsequent pathogenetic phenomena.
Although neurotoxicity of Aβ has been initially attributed to its fibrillar forms, more recent studies showed that neurotoxins also comprise small diffusible Aβ oligomers called Aβ-derived diffusible ligands (ADDLs), which were found to kill mature neurons in organotypic central nervous system (CNS) cultures [63]. At cell surfaces, ADDLs bound to trypsin-sensitive sites and surface-derived tryptic peptides blocked binding and afforded neuroprotection. Remarkably, neurological dysfunction evoked by ADDLs occurred well in advance of cellular degeneration. Recently it has been demonstrated that non-fibrillar assemblies of Aβ possess electrophysiological activity, with the corollary that they may produce dementia by disrupting neuronal signalling prior to cell death [64]. Recent experiments have detected the presence of ADDLs in AD-afflicted brain tissue and in transgenic mice models of AD [65–67]. The presence of high affinity ADDL binding proteins in hippocampus and frontal cortex but not cerebellum parallels the regional specificity of AD pathology and suggests involvement of a toxin receptor-mediated mechanism. The properties of ADDLs and their presence in AD-distressed brain are consistent with their putative role even in the earliest stages of AD, including forms of mild cognitive impairment. Considering the central role of Aβ in AD pathology, Aβ peptides and mechanisms for their formation have been the primary target in a number of contemporary attempts to develop therapeutic agents against AD.
Complement-mediated neurodegeneration
Although viewed for years as a so-called immuno-privileged organ, the CNS contains and synthesizes many components of the immune system. Both glia and nerve cells in the brain can synthesize immunoglobulins [68] and complement components [69, 70]. Brain cells can express all of the complement components and should thus be considered as an important source of complement [71].
The two major fibrillar amyloid forms Aβ40 and Aβ42 directly activate the alternative and classical pathways of the complement system by binding to C3 and the globular B head of C1q resulting in the formation of pro-inflammatory molecules such as C3a, C5a and membrane attack complex (MAC). MAC permeabilizes the cell membrane, which can result in cell lysis (Fig. 2). Astrocytes and microglia in the CNS produce both soluble complement regulatory proteins (CReg) and membrane-bound CReg (CD59, CD55, CD46) [72], which ensures a good level of protection against complement lysis by inhibiting MAC formation. However, the same cannot be said for neurons. There is very little information regarding the expression of CReg in neuronal cells. Studies to date suggest that cultured primary neurons express only low levels of the membrane CReg (mCReg), CD59 and CD55 [73, 74]. Furthermore, in the frontal cortex and hippocampus of AD brains, a significant decrease in the expression of CD59 at both protein and mRNA levels was found when compared with brains of age-matched control individuals without dementia [75]. An interesting hypothesis suggests that at sublytic concentrations, MAC can alter the function of neurons by triggering various cellular signalling pathways [8]; however, this needs to be further explored and corroborated in further systematic investigations.
Figure 2.

Activation of complement system by the amyloid plaques and its dual role in neuronal death. C1q binds to the boxed sequence in aggregated amyloid peptides and activates classical pathway of complement system. This leads to cleavage of C3 to C3b which could bind to a different sequence from the amyloid peptide (boxed). C3b initiates generation of C5-convertase cleaving C5 into C5a and C5b. C5a interacts with the C5a receptor (C5aR) on the surface of neurons and activates the neuroprotective mitogen activated protein kinases. On the other hand, C5b initiates the terminal pathway of complement cascade which forms MAC which integrates into membrane of the weakly protected neurons resulting in their death.
There is recent evidence that complement may also play a protective role in AD brain. The most convincing data for the protective role of components of the complement cascade demonstrate that production of C5a results in an activation of the neuroprotective mitogen activated protein kinases [76] (Fig. 2). Furthermore, animals genetically deficient in the complement component C5 were found to be more susceptible to hippocampal excitotoxic lesions [77]. These findings suggest a novel non-inflammatory role for C5a in modulating neuronal responses to excitotoxins. More recently, the protective role of the early activation stages of the complement cascade in AD was demonstrated in transgenic mice with human APP, in which complement was blocked by expression of soluble Crry (a rodent CReg with inhibitory activities similar to human CD46 and CD55) [78]. Results showed that mice expressing soluble Crry had a 2–3-fold increase in Aβ accumulation and neuronal degeneration compared to animals that did not express the inhibitor. However, Crry inhibits C3 activation, and thus prevents generation of C5a. It was suggested that the observed effect might be due to decreased C5a formation. Another recent study found that C1q protected cultured primary neurons against Aβ and SAP (serum amyloid P) induced neurotoxicity [79]. The exact mechanism of this protection has not yet been elucidated but the data suggest that C1q does not protect from Aβ induced apoptosis. However, considering the protective role of C5a, the C1q-mediated defence against senile plaques might be at least in part due to the generation of C5a. This speculation is supported by a recently published in vivo study on C3 deficient APP transgenic mice that demonstrated accelerated plaque deposition and neurodegeneration compared to C3 sufficient animals [80].
Although it is still disputable if complement system is a primary cause of neuronal degeneration or a secondary event in AD [81], the findings discussed above demonstrate that complement not only lyses neurons, but also generates inflammation resulting in an amplification of AD risk factors and neuronal death [17]. Thus, targeting the complement cascade seems an attractive candidate strategy for treating AD and other neurodegenerative diseases. However, complement seems to possess a dual, ambiguous role in the pathogenesis and progression of the AD which needs to be carefully considered when choosing targets for therapeutics to be developed.
Oxidative and nitrosylative damage
This hypothesis emphasizes the role of reactive oxygen and nitrogen species (ROS or RNS) in initiation and promotion of neurodegeneration in the brains of AD patients [18, 19]. Some of these radicals originate from normal brain function processes: it is known that the brain utilizes about 25% of respired oxygen and that about 2% of the oxygen consumed converts into ROS. Other free radicals are released during inflammatory reactions and by Aβ. This hypothesis for the AD aetiology is supported by the observation in some clinical trials that high doses of antioxidants might be beneficial to AD patients [82, 83].
Increased oxidative stress may enhance intracellular accumulation of Aβ in neurons [84]. In addition, studies show that membranes containing oxidatively damaged phospholipids accumulated Aβ faster than membranes containing only normal saturated phospholipids [85]. Based on this finding, it was proposed that one or some of the mechanisms of action in Aβ neurotoxicity might be free-radical mediated [86]. Further studies on protection of cultured neuronal cells against Aβ-induced toxicity by vitamin E supported this hypothesis [87]. Additional evidence of increased oxidative stress in AD include: AD patients have lower levels of vitamins A, E and β-carotene in their serum compared to control patients [88]; increased consumption of oxygen is found in AD patients [89]; activation of calcium-dependent neural proteinase (calpain), triggering formation of free radicals, was found in AD autopsy brain samples [90]; homogenates of frontal cortex from such autopsy samples revealed a 22% higher production of free radicals than those of age-matched controls [91]; increased neuronal nitric oxide synthase (nNOS) expression in reactive astrocytes correlated with apoptosis in hippocampal neurons of post-mortem brains from AD patients [92]; glutamine synthetase, a highly sensitive enzyme to oxidative stress, showed decreased activity in the AD autopsy brains [93]; increased levels of oxidized proteins are found in the blood of AD patients when compared with non-AD controls [93].
Proteasome inhibitor-induced neurotoxicity
Numerous studies now clearly demonstrate that inhibition of the proteasome is sufficient to induce cell death in both neuronal and glial cells. These studies have shown that proteasome inhibitors can induce hallmarks of apoptosis, including caspase activation, cytochrome C release, elevated p53 expression, chromatin fragmentation and DNA laddering [94–99]. The induction of such a wide variety of cell death events by proteasome inhibitors suggests a critical role for proteasome activity in neuronal homeostasis. The proteasome is directly responsible for the degradation of a number of cell death factors and its inhibition is sufficient to increase their levels to a cytotoxic level. Furthermore, the proteasome prevents accumulation of proteins not directly related to cell death, but which can contribute to neuron death by conferring an increased half-life of neurotoxic proteins. For example, proteasome inhibition has been shown to increase the accumulation of cytotoxic Aβ[100, 101] and presenilin [102, 103]. It has been shown that heat shock proteins (HSPs) can enhance proteasome-mediated proteolysis [104, 105].
However, proteasome inhibition does not induce neuronal death in all neuron populations or experimental setups [106, 107]. It seems that proteasome inhibitor-induced toxicity is cell-type specific. It was demonstrated that proteasome inhibition could have very different effects on cell death based on the differential role of NF-κB – a downstream proteasome target which can have pro-apoptotic or anti-apoptotic effects depending on cell type [108, 109]. In transferring the knowledge obtained from in vitro studies to studies in the mature CNS, it is important to consider the fact that the majority of in vitro studies are conducted in cultures established from embryonic tissues or early postnatal brain. One thus must take into account the possibility that embryonic tissue may have a different dependence on proteasome activity than established neurons within the developed and mature, or, as in the case of AD patients, aged CNS.
Only recently, the importance of the proteasome in the physiology and pathology of the CNS has begun to emerge. It is likely that studies on this intriguing enzyme will yield further important information on how oxidative stress occurs in the CNS and how CNS protein turnover is regulated. Ultimately, this may lead to the design of useful new therapeutic strategies aimed at eliminating oxidative stress toxicity in the CNS, thereby contributing to the prevention and/or amelioration of clinical AD symptoms.
Cholesterol-induced neurotoxicity
All mammalian cells require cholesterol for the formation and maintenance of cell membrane permeability and fluidity. There is increasing evidence in support of the hypothesis that alterations in cholesterol levels influence the development of AD by affecting Aβ formation and distribution within cholesterol rich membranes. Proteolytic processing of APP is believed to occur at or in close proximity to the cholesterol-rich plasma membrane. When cultured human cells transfected with the APP cDNA are exposed to an inhibitor of the HMG-CoA reductase (the rate controlling enzyme of the metabolic pathway that produces cholesterol and other isoprenoids) β-secretase cleavage of newly synthesized APP is markedly reduced. Addition of solubilized cholesterol to these cells causes a four-fold increase in newly synthesized β-amyloidogenic products, while reduction of the cellular cholesterol level of hippocampal neurons inhibits formation of Aβ[110].
Above in vitro data correlate well with both in vivo experiments using AD animal models and ex vivo data obtained from post-mortem brains of AD patients. In a double transgenic mouse that overexpresses mutant APP and presenilin-1, a high cholesterol diet resulted in significantly increased levels of Aβ in brain tissue. The number and size of Aβ deposits were elevated. Levels of total Aβ were strongly correlated with the levels of both plasma and CNS cholesterol [111]. When these mice were treated with an inhibitor of HMG CoA reductase, a cholesterol-lowering drug, plasma cholesterol, brain Aβ peptidesand Aβ load decreased by more than two-fold [112]. Results in cholesterol-fed rabbit models are consistent with these results in murine transgenic models. Cholesterol-fed rabbits produce and accumulate Aβ in the brain and this accumulation can be reversed by removing cholesterol from the rabbits’ diet [113, 114]. In post-mortem brains of AD patients, fluorometric staining has confirmed cholesterol accumulation in the core of mature senile plaques. Although it is not firmly established whether disruption of cholesterol balance at the cellular level is a primary cause of AD or rather a secondary effect of the disorder, the above evidence strongly support the view that membrane cholesterol is responsible for the binding and toxicity of Aβ.
Viral infections
Although the exact mechanisms of driving neuronal death by viral infection are not very clear, it was proposed that string inflammatory responses promoted by viral infections might be responsible for the increased risk of developing AD. A comparative examination of post-mortem brain tissue from 97 people with HIV/AIDS and 125 non-infected people of similar age established that Aβ plaques, indicative of AD, generally increased with age in both groups, but were more than twice as common in the HIV/AIDS group. Almost one in five of those who had died of AIDS in their 40s had the characteristic plaques but there was no evidence these plaques in the brain tissue of non-infected people of the same age. Two out of the ten HIV infected patients in their late 30s and early 40s who had died before developing AIDS also had the characteristic plaques. These findings strongly indicated that an inflammatory response in the brain, such as that caused by viral infection, may initiate and/or accelerate the pathological processes leading to AD [115].
Actually, a number of viral or bacterial pathogens (Chlamydia Pneumoniae, Herpes simplex type 1 (HSV-1) and type 2 (HSV-2), herpes virus 6 (HHV-6), cytomegalovirus (CMV) and Helicobacter pylori bacterium) have been implicated in AD [116–118]. As these pathogens are so common, however, they probably only contribute to AD if acting in concert with other factors, or under certain specific conditions. It was shown that the risk-promoting effects of Herpes simplex in AD are influenced by a number of molecular mechanisms such as possession of the ApoE4 allele [119], APP processing [120], and possible involvement of APP in the intraneuronal traffic of the virus [121]. Herpes infection was shown to increase the production of Aβ in mouse brains [122]. Numerous other genes and proteins implicated in AD interact with the products of the Herpes simplex viral genome or play a role in its life cycle, suggesting a complex synergy between host and pathogen that may play an important role in the pathophysiology of AD (reviewed in [3]).
Interrelationship between different aetiopathogenic theories of AD
Many possible risk factors for AD have been investigated, with only very few showing positive associations and none defining the aetiology of the neurodegenerative disease on its own. Therefore, it is likely that interplay between at least some of these potential risk factors determines AD pathogenesis via a pathophysiological cascade.
Substantial evidence suggests that, apart from age, a history of head injury [123–127] and/or certain types of viral infections [128–131] increase the risk of AD. These two risk factors, however, are accompanied by a chronic inflammatory reaction leading to the release of a number of cytokines, including interferon (IFN)-γ and TNF-α. An inhibitory effect of IFN-γ on the translational machinery has been well-documented [132–134], leading to lower expression of proteins, including SC35 and hnRNPA1, whose downmodulation results in increased secretion of Aβ by cultured neurons [135]. A cytokine-dependent enhancement of Aβ production from APP-expressing astrocytes and cortical neurons in transgenic mice was also demonstrated [136]. Furthermore, HSV-1 protein ICP27 has been shown to inhibit host cell splicing [137–139] by altering phosphorylation of SC35 and other proteins [140], which we showed to yield in accumulation of Aβ[135]. Cholesterol and other ‘raft’ lipid components, such as sphingomyelin and galactosylceramide, induce aggregation of the secreted Aβ peptide [141, 142].The accumulated amyloid plaques on the other hand cause neuronal death by triggering a number of processes such as generation of ROS and RNS and inhibition of proteasomes, but one of the most important mechanisms is the activation of the complement system that not only lyses neurons, but also generates inflammation which in turn amplifies the AD risk factors. Due to the exceptionally complex events involved in AD neurodegeneration and their reciprocal relationship and interaction, it is difficult to determine what the primary cause of the disease is. It is likely that different primary risk factors synergize to initiate pathology. It is also possible that several different initial pathomechanisms ultimately result in a similar neuropathology (plaques, tangles) and clinical phenomenology (dementia), thus raising the question whether the diagnosis of AD really represents a single nosological entity.
Treatment approaches
AD is a very complex disease and its clinical management is highly challenging. Personality and behavioural changes, and the progressive difficulties with performing activities of daily living lead to dependence on external help and support. A cure for AD is still not available and clinicians and caregivers are challenged by an increasingly aging population with ever higher risk of dementia. The primary goals of treatment are to maximize the patient’s ability to function in daily life, maintain quality of life, slow the progression of symptoms, treat depression and disruptive behaviour. However, at present all treatments for AD offer only modest symptomatic relief for periods of between six to eighteen months. Some of the currently used/researched agents are considered to delay or even prevent AD symptoms which could contribute to reducing the number of patient with dementia. However, until now complement proteins and CRegs have been overlooked as targets for the development of neuroprotective drugs for AD treatment. But accumulated data suggest that appropriate modulation of expression and/or activity of proteins controlling the complement cascade could be very beneficial for AD patients. Here we review the current, albeit still unsatisfactory state of this field. There are a number of different categories of drugs currently used in AD treatment (summarized in Table 1) and examples of each group are discussed below. Based on the ongoing research we also speculate on the future developments regarding AD therapeutics.
Table 1.
| Agents | Targets/mechanisms | Status |
|---|---|---|
| Disease-modifying therapies | ||
| Donepezil (Aricepts) | Cholinesterase inhibitor | FDA approved |
| Rivastigmine (Exelon) | Cholinesterase inhibitor | FDA approved |
| Galantamine (Reminyl) | Cholinesterase inhibitor | FDA approved |
| Physostigmine Salicylate (Synapton) | Acetylcholinesterase inhibitor | Discontinued |
| Milameline (CI 979) | Partial muscarinic agonist; Increases central cholinergic activity | Discontinued |
| AF 102B (cevimeline HCL, Evoxac) | Blocks production of Aβ by increasing the activity of α-secretase and possibly by inhibiting γ-secretase | Discontinued |
| SB202026 (Memric, Sabcomeline) | Selective muscarinic M1 partial agonist | Discontinued |
| Tacrine (Cognex) | Acetylcholinesterase inhibitor | FDA approved |
| Metrifonate | Cholinesterase inhibitor | Discontinued |
| Memantine (Ebix, Namenda) | NMDA receptor antagonist | FDA approved |
| LY450139 | γ-secretase inhibitor | Phase III |
| PF-04494700 (TTP488) | Inhibitor of Receptor for Advanced Glycation Endproducts (RAGE) | Phase II/IIa/IIb |
| NGX267 (AF267B) | M1 muscarinic agonist, α-secretase activator | Discontinued |
| AZD3480 (ispronicline, TC-1734) | Nicotinic agonist with high selectivity for neuronal α4β2 nicotinic (nAChR) receptors | Phase II/IIa/IIb |
| BMS-708163 | γ-secretase inhibitor | Phase II/IIa/IIb |
| AL-108 | Stabilizes microtubules, blocks Aβ aggregation | Phase II |
| MK 0752 | γ-secretase inhibitor | Phase II |
| E2012 | γ-secretase inhibitor | Phase I |
| AZD103 ATG-Z1 | Inhibits Aβ fibrillization and disassembles preformed amyloid fibrils | Phase I Preclinical |
| OM99-2 | BACE1 inhibitor | Investigational |
| KMI-429 | Analogue for the aspartyle protease, BACE1 inhibitor | Investigational |
| GRL-8234 | Analogue for the aspartyle protease, BACE1 inhibitor | Investigational |
| KNI-1027 | BACE1 inhibitor | Investigational |
| GSK188909 | BACE1 inhibitor | Investigational |
| BACE1 inhibitor | ||
| Symptom-management therapies | ||
| Alpha-tocopherol (Vitamin E) | Phase III | |
| Acetyl-l-carnitine HCI (ALCAR) | Destroys toxic free radicals Not exactly known (some hypotheses: mitochondrial/energy | Discontinued |
| Cerebrolysin | production; stabilizes membranes; decrease accumulations of toxic fatty acids) | FDA approved (outside USA) |
| Ibuprofen | Exerts nerve growth factor like activity on neurons from dorsal root ganglia; neurotrophic and neuroprotective agent | Phase III |
| Flurizan (MPC-7869, r-flurbiprofen) | Reduces prostaglandin activity by inhibiting prostaglandin | Discontinued |
| Huperzine (Cerebra capsule, Pharmassure) | synthetase; anti-inflammatory, analgesic, and some antipyretic activity Selective amyloid-lowering agent | Phase II/IIa/IIb Discontinued |
| Naproxen (Aleve, Anaprox, Naprosyn) Estrogen (Premarin) | Selective inhibition of AChE; alterations in APP processing; reduction of neurotoxicity by Aβ; antioxidant effects; increase of NGF production | Phase III Phase III |
| Xaliproden | Anti-inflammatory | Phase III |
| Lecozotan Dimebon | Thought to enhance growth of neurons in the basal forebrain; may have a direct effect on Aβ 5-HT1a receptor agonist | Phase III Phase II/IIa/IIb |
| PRX-03410 | 5-HT1a receptor antagonist | Phase II/IIa/IIb |
| MEM 1003 | Inhibitor of cholinesterase and NMDA receptors; Inhibits neuronal death, potentially by mitochondrial-mediated inhibition of apoptosis | Phase II/IIa/IIb |
| MEM 3454 (RO5313534) | Partial 5-HT4 receptor agonist | Phase II/IIa/IIb |
| CERE-110 (Nerve Growth Factor Gene Therapy) | Neuronal L-type calcium channel antagonist Selective nicotinic alpha-7 receptor partial agonist; 5-HT3 receptor antagonist | |
| Inhibitors of complement system Naturally occurring | Trophic agent in the survival and maintenance of basal forebrain cholinergic neurons | |
| Polyphenolic flavonoids | Investigational | |
| β-glycyrrhetinic acid steroid-like | Investigational | |
| Polysaccharides (GR-2II, AGIIb-, BR-5I, AR-2IIa) | Investigational | |
| Sulfated polysaccharide (fucan) | C3/C5-convertase inhibitors Inhibitor of Classical pathway | Investigational |
| Esters (rosmarinic acid) | In vitro inhibition of complement cascade at different stages | Investigational |
| Polyanionic carbohydrates | Investigational | |
| Alkaloids | Inhibit the classical pathway by interfering with C1 activation or by inhibiting | Investigational |
| Ca2+-binding polymers | C3 cleavage | Investigational |
| Glycoproteins (CI, CVF) | C3/C5-convertase inhibitors | Investigational |
| Peptide-like related to glycopeptide antibiotics (complestatin) | Inhibitors of classical and alternative pathways Inhibitors of classical pathway | Investigational |
| Fungal metabolite K-76 | Inhibitors of classical pathway | Investigational |
| Proteoglycans (decorin) | C3-convertase inhibitors | Investigational |
| Glycosaminoglycans (heparin) | Inhibitors of classical and alternative pathways | Investigational |
| Chondroitin sulphate proteoglycan (GCRF, CSPG) | Inhibitors of classical and alternative pathways | Investigational |
| C4-binding protein (C4bp) | C1q inhibitor | Investigational |
| Cyclic hexadepsipeptides | C3-convertase inhibitor C3bBb, factor B, C1q inhibitors | Investigational |
| Synthetic inhibitors | Investigational | |
| Peptide analogues and derivatives | C4 inactivation in vitro and in vivo | |
| (C089, PR226, CBP2, compstatin, etc.) | C5aR antagonist in vitro and in vivo | Investigational |
| Diisopropyl fluorophosphates (DFP, BCX-1470) | C5aR or C3aR antagonists, bind to either C3, factor D, factor B or C1q | Investigational |
| K-76 analogues (TKIXc, K-76COOH) | Investigational | |
| Nafamstat mesilate (FUT-175) | Investigational | |
| Oligodeoxyribonucleotide containing phosphorothioate (PS-oligo) | Factor D binding | Investigational |
| Triterpenoid oleanolic acid derivatives | Inhibitors of classical and alternative pathways | Investigational |
| Inhibitory prodrug proteins (CD55- and CD59-prodrugs) | Inhibitors of classical and alternative pathways Inhibitors of classical and alternative pathways | |
| C3-convertase inhibitor Inhibition of C3/C5-convertases or membrane attack complex (MAC) | ||
Drugs which modify AD symptoms
Cholinergic treatment
Acetylcholinesterase inhibitors are one of the most commonly used drugs to treat AD [143]. Their use stems from the observation that there is a deficiency in cholinergic neurotransmission in AD. According to one hypothesis, decreased cholinergic transmission plays a major role in the development of cognitive, functional and behavioural symptoms of AD [144, 145]. As a result of the pathological changes during AD there are decreases in biochemical indices of cholinergic functioning in neocortex and hippocampus that correlate with dementia severity [146–148]. The hypothesis is further supported by pharmacological studies using cholinergic agonists and antagonists and transgenic animal models that emphasize the close connection between cognition and cholinergic neurotransmission [149]. The cholinesterase inhibitors currently approved by the US Food and Drug Administration (FDA) are tacrine, donepezil, rivastigmine and galantamine. The mechanism of action of these drugs consists in increasing the availability of acetylcholine through an inhibition of the catabolic enzyme acetylcholinesterase.
Antiglutamic treatment
Another approach is to block glutamic neurotransmission. Glutamate is the main excitatory neurotransmitter in the brain and has been implicated in so-called excitotoxicity. One of its receptors – M-methyl-D-aspartate (NMDA), has been implicated in long-term potentiation, the neuronal mechanism responsible for learning and memory [150]. The low affinity non-competitive NMDA antagonist, Memantine, was approved by the FDA for AD treatment in 2004. Memantine acts by blocking the NMDA receptor, thus preventing calcium influx when neuronal firing rates are high, but leaves the calcium channel open for transmission at low stimulation rates [151]. Clinical studies demonstrate that the drug has rather symptomatic benefits for patients than a disease modifying effect [152, 153].
β- and γ-secretase inhibitors
β and γ secretases are the two enzymes critically responsible for the formation of amyloid plaques (Fig. 1). Therefore, based on the amyloid hypothesis, drugs that can prevent production, aggregation and deposition of Aβ are thought to be promising therapeutics for AD. In particular, the amyloidogenic pathway in neurons is initiated by β-secretase cleavage. The elucidation of the molecular identity of β-secretase, also called BACE1 (β-site APP cleaving enzyme 1) had been one of the key issues for the development of AD therapeutics. Animal experiments showed that mice lacking β-secretase are fertile, do not produce Aβ42 and have no obvious neurological deficits [154]. Clinical data support this approach since activity of β-secretase increases with age [155]. Development of β-secretase inhibitors (BSI), however, has proven to be challenging and to date none has been tested extensively in human beings [156]. The crystal structure of BACE1 shows that the active site is comprised a long cleft for substrate recognition [157]. The structural features of BACE1 are used for design of BSI. The first generation of BSI were designed as transition-state analogues for the aspartyle protease (e.g. OM99–2, KMI-429). The majority of commercially available potent BSI are peptide-based compounds containing a transition-state moiety (e.g. hydroxyethylene, statin, etc.). Promising recent data suggest that KMI-429, has great potential as it significantly reduced production and accumulation of senile plaques in vivo in brains of wild-type and transgenic mice [158]. A new generation of BSI was designed recently [159], containing hydroxyethylamine isostere and isophthalamide moiety (GRL-8234). This compound efficiently inhibits BACE1 activity not only in vitro but also in vivo. Another recently generated BSI containing a 2,6-pyridinedicarboxylyc, chelidamic or chelidonic residue at the P2 position together with hydroxymethylcarbonyl isostere (KNI-1027) was also shown to inhibit efficiently BACE1 [160]. Considering the usage of BSI as drugs, making less peptidic compounds is mandatory to obtain sufficient oral absorption and penetration through the blood-brain barrier. These two issues have hampered progress in development of BSI for AD therapeutics, however, recently was generated the first orally bioactive non-peptidic BSI, GSK188909, able to reduce brain Aβ levels in APP transgenic mice [161].
Although γ-secretase has in many ways been an attractive target for AD therapeutics, interference with Notch processing and signalling may lead to toxicities that preclude clinical use of inhibitors of this protease. Knockout of Notch1 or PS1 is embryonic-lethal in mice [162, 163] but Notch signalling and γ-secretase activity are crucial in adulthood as well, because Notch plays a critical role in many cell differentiation events [164]. Indeed, treatment of mice with γ-secretase inhibitor (GSI) LY-411575 at 10 mg/kg/day for 15 days caused severe gastrointestinal toxicity and, at 10 mg/kg/day, also interfered with the maturation of B and T lymphocytes, effects that are due to inhibition of Notch processing and signalling [165, 166]. The related compound, benzolactam LY-450139, is two orders of magnitude less potent compared to LY-411575; however, this compound has moved into clinical trials, and so far it is the only GSI for which human trials have been reported. LY-450139 was chronically administered for 5 months to young APP transgenic mice, leading to reduced total brain Aβ and slower formation of Aβ plaques. In initial human trials in healthy volunteers [167] single doses of this GSI of up to 140 mg were apparently safe and reduced plasma Aβ levels by up to three quarters. However, steady-state Aβ in the CSF was not affected, and it is unclear if higher doses can lower brain Aβ without Notch-related side effects. Sulphonamide inhibitor BMS-299897 was developed to be selective for inhibiting the processing of APP over Notch [168, 169], although the use of different assays in this study does not allow simple comparisons. Single oral administration of this compound into APP transgenic mice gave an ED50 value of 30 mg/kg for lowering brain Aβ and 16 mg/kg for lower plasma Aβ at 3 hrs after dosing. Intraperitoneal administration in guinea pigs, which naturally produce high levels of Aβ that is identical in sequence to the human peptide, reduced brain, plasma and CSF Aβ with an ED50 of 30 mg/kg at 3 hrs after dosing.
The hope of using GSI for treatment of AD is tempered by the fact that γ-secretase cleaves numerous other type I membrane protein stubs that result from ectodomain shedding [170]. Agents selective for APP versus Notch may reveal new long-term toxicities due to blocking proteolysis of these other substrates, toxicities masked by the severe Notch-related effects with non-selective inhibitors. To address this important issue, the development of more potent agents that work by this mechanism will be critical. Further detailed analysis of design and therapeutic potential of BSI and GSI could be found in recently published reviews [171, 172] dedicated specifically to this issue.
Drugs which potentially prevent and modify AD symptoms
Vitamins and antioxidants
Growing evidence supporting the oxidative stress hypothesis has led to the concept of using antioxidants such as vitamin E (α-tocopherol) as a potential treatment approach in AD. However, vitamin E in combination with selegiline, a drug used for the treatment of early-stage Parkinson’s disease, depression and senile dementia, showed no real benefits for the patients [82]. Other clinical trials also failed to show beneficial effects of vitamin E administration to AD patients [173–175]. These results together with recent observations that high doses of vitamin E (>400 IU/day) may increase mortality [176] suggest that vitamin E should not be given as an additive to other therapeutic approaches for AD treatment. Clinical results for selegeline treatment have been rather controversial suggesting either only a small benefit or no significant improvement in the cognition of AD patients [177].
The extract of the plant Ginkgo biloba was suggested to enhance cognition. In addition to putative antioxidant properties, the extract has been reported to reduce the aggregation of the Aβ peptide. However, in a randomized trial Ginkgo biloba did not significantly enhance memory in healthy elderly adults [178].
Non-steroidal anti-inflammatory drugs
Retrospective observational studies have shown that the use of NSAIDs may have protective properties regarding the development of AD [10, 179–182]. In epidemiological studies, the use of NSAIDs was associated with lower risk of developing AD and the benefits seem to be greater with long-term use [179]. However, NSAIDs only seem to offer protection if used before disease onset, treatment after the development of AD symptoms results in little or no benefit to the patients [183–187]. Therefore, currently NSAIDs are not used in the treatment of AD.
Oestrogen replacement therapy
Women have two- to three-fold higher age-specific prevalence rates of AD [188]. Women are oestrogen deficient after menopause, whereas men rather maintain hormone levels as testosterone undergoes aromatization to estradiol. Oestrogen is considered to have a number of beneficial features such as antioxidant and anti-inflammatory properties, interactions with neurotransmitters such as acetylcholine, and an ability to alter apolipoprotein which could lower the risk of developing AD. In vivo studies with low doses of conjugated oestrogen (Premarin) in a rat model showed that this treatment can prevent vascular deposition of Aβ, thus protecting the endothelial cell from toxic effects of the plaques [188, 189]. An anti-inflammatory effect of the Premarin has also been demonstrated.
Inhibitors of complement-mediated degeneration
Recently we reviewed in detail the important dual role of the complement system and its regulators in brain inflammation and neuronal damage in AD [17]. This dual role should be carefully considered when choosing therapeutic targets for AD treatment. Until now, complement proteins and CRegs have been largely overlooked as targets for developing AD drugs, but accumulated data suggest that appropriate modulation of expression and/or activity of proteins controlling the complement cascade could be of great clinical benefit for AD patients.
Cobra venom factor (CVF)
The earliest indication that inhibition of complement might be of therapeutic benefit came from analyses of the effects of decomplementation with CVF [190–194]. CVF binds mammalian fB in plasma resulting in cleavage of fB by fD and production of C3 convertase [195]. This process leads to consumption of all plasma C3 and lack of functional complement. However, after a week treatment with CVF, experimental animals develop neutralising antibodies against CVF which makes it ineffective for longer-term therapeutic applications, e.g. in AD. Nevertheless, CVF has provided a useful proof-of-concept for the theory that complement activation is of relevance in diverse neuropathologies.
Heparin and other polyionic agents
Heparin, a polyanionic glycosaminoglycan, has long been recognized as an in vitro inhibitor of complement [196]. Heparin affects complement activation at multiple levels, by binding and inactivating C1, blocking generation of the C3 convertases and interfering with the assembly of MAC [197–200]. Very few studies have addressed the potential use of heparin as a complement inhibitor in vivo. N-acetylated heparin with much reduced anticoagulant activity can inhibit CVF-induced complement activation in guinea pigs [201]. Further in vitro work on human samples demonstrated that heparins were much more efficient inhibitors in human than in guinea pig serum [202]. Interestingly, it was recently demonstrated that this drug can interfere with the metabolism of amyloid plaques and reduces the accumulation of Aβ[203]. Long-term treatment with enoxaparin, a low molecular weight heparin, in human APP751 transgenic mice can significantly reduce the deposition of amyloid plaques and Aβ aggregation [203]. Chronic treatment with enoxaparin is well tolerated and does not increase side effects such as haemorrhage in the brain. Although the exact mechanism is not yet clear, a reduced ability of Aβ to activate complement system and its classical pathway may play a role.
Numerous related and unrelated polyanionic molecules, including dextran sulphate, polyvinyl sulphate, polylysine and suramin, inhibit complement activation in vitro (reviewed in [204]). However, their applicability in vivo and particularly for treatment of AD has not been investigated.
Other small molecule inhibitors
Many synthetic and natural molecules have been shown to inhibit complement activation in vitro. A few natural inhibitors have shown efficient complement inhibition both in vitro and in animal models. The molecule termed K-76COOH, isolated from cultures of the fungus Stachybotrys complementi, inhibits complement at the C5 stage [205, 206]. Rosmarinic acid, extracted from the common shrub Rosemary, covalently binds activated C3b and prevents formation of the convertase [207, 208]. An extract of the Chinese medicinal herb, Ephedra, inhibits complement at multiple levels including the terminal pathway [209]. Among the synthetic complement inhibitors, the protease inhibitor FUT-175 is one of the best investigated. It inhibits C1r, C1s, fD and the C3/C5 convertases [210, 211]. FUT-175 is effective in numerous animal models of complement-mediated disease [212, 213], although it has not been studied in AD models. Some success has also been obtained using small molecules, such as peptides, that bind active sites of complement components or enzymes and prevent their participation in the cascade. Examples of such peptides are C089, PR226, CBP2 and compstatin. The use of the small molecule inhibitors, however, is currently hampered by non-specific side effects or short half-life [214] and therefore, none of them are likely to be useful for therapy of chronic diseases such as AD unless these problems can be solved.
Recombinant protein inhibitors
In addition to the approaches described above, a plethora of soluble therapeutics has been developed which specifically inhibit the complement cascade [215]. Early agents were based on the naturally occurring membrane-associated CRegs, such as CD49, CD59 and Complement Receptor 1(CR1; CD35). Removal of membrane-anchoring domains using recombinant technology generated soluble molecules with the same regulatory functions as the ‘parent’ inhibitor which could be administered locally or systemically to inhibit complement. The best known of these is a soluble form of CR1 (sCR1), an agent developed nearly 30 years ago and used in the clinic, with limited success, for complement inhibition in myocardial infarction (MI) and cardiopulmonary bypass (CPB) [216–218]. These ‘first-generation’ agents have been engineered and modified over the years to improve characteristics such as half-life (by introducing antibody Fc domains) [219], membrane localisation (by using lipid ‘tails’) [220] and specific targeting of complement activation sites (by using fusion proteins comprising a domain that binds C3 activation fragments, thus localising to the site of C attack) [221]. Some success has also been obtained using small molecules, such as peptides, that bind active sites of complement components or enzymes and prevent their participation in the cascade. Their use, however, is hampered by non-specific side effects or short half-life [214]. Perhaps the most successful soluble therapeutic agents have been antibody-based agents, such as full-length immunoglobulin of single-chain Fv (scFv) fragments. Antibodies that bind C5, thereby preventing triggering of the terminal pathway and MAC formation, have been successfully used in the treatment of various pathologies exacerbated by complement activation. Anti-C5 in particular is used for treatment of paroxysmal nocturnal haemoglobinuria, MI and arthritis [222–224].
The problem however with many of these agents is that their action is systemic, totally blocking the complement cascade. In addition, if they are targeting the alternative, classical or MBL activation pathways, their long-term use can be detrimental, triggering immune complex disease and permitting bacterial infections as the key functions of complement in innate immunity are prevented. In the case of AD, it may be beneficial to selectively inhibit parts of the complement cascade. For example, inhibition of the terminal pathway using antibodies that allow C5a formation but prevent MAC formation may prevent neuronal damage but still permit the observed protective effects of C5a. Another agent, developed to selectively inhibit MAC formation, but which will still allow C5a generation, is a CD59-Ig fusion protein [225]. This has been tested in a murine model of laser-induced choroidal neovascularization and shown to have therapeutic effects. CD59-Ig, which is a long-lived agent, halts the complement cascade at the level of C8. Agents that specifically target complement inhibition to the vicinity of the amyloid plaques would avoid systemic inhibition of C, and provide enhanced protection to neurons. Previous in vitro studies have demonstrated that recombinant techniques can be used to fuse CReg to the carboxy-termini of antibodies or antibody fragments, thus locating the therapeutic action of the inhibitor to the site pre-determined by antibody specificity [226]. If recombinant antibodies to Aβ, or other antigens unique to the plaque, are developed, then specific targeting of CD59 or other substrates at sites of disease may be possible. It is clear that the exact role of each complement component in AD is yet to be understood (see Complement-mediated neurodegeneration section). Only then, specifically targeted anti-complement agents will be likely to be developed and to be of therapeutic benefit. Testing prototype agents in murine models of AD, or further investigation of the role of complement in the disease using component-deficient animals will help clarify the potential of anti-complement therapy in AD.
Anti-amyloid therapy (‘vaccination’)
The major aim of anti-amyloid therapy is to induce clearance of the amyloid plaques, one of the defining risk factors and pathological hallmarks of AD. A largely unexpected discovery showed that immunization with Aβ42 peptide prevents the appearance of amyloid pathology in a transgenic mouse model of AD [227]. Follow-up studies confirmed this finding in other models of the disease [228]. These experiments suggested that passive immunization with antibodies against human Aβ should decrease the neuritic plaques and this hypothesis was confirmed in transgenic mice, including improved performance in behavioural tests [229, 230]. Further to these promising experiments in animal-models without apparent side effects, clinical trials with a vaccine designated as AN1792 containing pre-aggregated Aβ42 and QS21 as an adjuvant were carried out. The design of this vaccine aimed to induce a strong cell-mediated immune response, since QS21 is a strong inducer of Th1 pro-inflammatory lymphocytes [231]. This initial trial involved 80 patients with mild to moderate AD [232] and was design to assess the antigenicity and toxicity of multiple immunisations with Aβ42 peptide containing QS21. Slightly more than half of the patients (53%) developed antibodies against Aβ. During the later stages of the phase I trial, the emulsifier polysorbate 80 was added to the vaccine, shifting the response from a predominantly Th2 biased to a pro-inflammatory Th1 [233]. In the subsequent phase II trial, 372 patients were enrolled with 300 receiving AN1792 in the polysorbate 80 formulation. Unfortunately, this trial had to be stopped prematurely after 6% of patients (18 out of 298 subjects) who received the vaccine developed acute meningoencephalitis [234]. Nevertheless, the trial and ex vivo studies yielded some important data. Striking Aβ clearance of parenchymal plaques had occurred, similar to what had been reported in the animal studies, confirming the validity of this approach for amyloid clearance in human beings [235–239]. Antibodies raised by this active immunisation were specific for senile plaques only and did not cross-react with the full-length APP or other derivates [240]. Furthermore, neuropathological evaluation suggested involvement of activated microglia in plaque clearance [239, 241]. Another interesting observation suggested a mechanism for the observed Aβ plaque clearance, which actively involves complement system. C1q enhances the uptake of antibody-fibrillar Aβ complexes in vitro by microglia, possibly via C1qRp receptor [242]. Despite the striking Aβ clearance, several significant problems were apparent from the pathological and clinical data from these trials. Some of the pathologically examined cases showed a harmful T-cell reaction surrounding cerebral vessels, suggesting an excessive Th1 immune response. It appeared that the immune reaction triggered by the AN1792 vaccine was a double-edge sward, where the benefits of a humoural response against Aβ42 were surpassed in some individuals by a detrimental Th1-mediated inflammatory response [235, 243]. Furthermore, despite the apparent success in amyloid clearance, the clinical cognitive benefits from the vaccination were very modest compared to the placebo group [244, 245]. The above observations suggest a possibly promising future for passive or active immunisation against amyloid plaques. However, further studies on optimal time to begin immunisation, design of improved vaccines and pre-clinical models, determination of most appropriate target for vaccination (detail discussed in [246]) are mandatory. A combination of anti-amyloid therapy with anti-inflammatory therapeutics might be a solution to overcome currently existing problems such as devastating side effects (meningoencephalitis). However, whether such a combination would be beneficial for AD patients needs to be addressed in further empirical studies, after its safety has been confirmed.
Another interesting approach is to prevent cleavage of APP in the first place by blocking the cleavage site of β-secretases and thus formation of Aβ[247]. Antibodies that block APP cleavage by β-secretase were tested in an in vitro model in which cells expressed wild-type APP. These antibodies bound full-length APP, internalized it into the cells and inhibited both intra- and extracellular Aβ peptide formation. The same antibody has also been tested in a transgenic mouse model expressing human APP. The animals showed improved cognitive functions and a reduction in brain inflammation as well as in the incidence of microhaemorrhage. Interestingly, these beneficial effects were not associated with the levels of Aβ which were unaltered by antibody treatment [247]. Therefore, the exact mechanism by which this antibody induced these improvements in transgenic animals, remains to be elucidated.
Targeting proteasome inhibition
As proteasome inhibition may play a contributing role to neurodegeneration, it is important to determine mechanisms which may modulate that inhibition. One obvious target for proteasome inhibition could be an efficient up-regulation of the levels of HSPs within the CNS. Increasing the concentration of molecules of the HSP family should help to preserve proteasome activity and possibly result in delaying age-related proteasome inhibition. Alternatively, pharmacological interventions designed to stimulate proteasome activity within the CNS may provide some therapeutic benefit. Different studies have identified a number of pharmacological proteasome activators that may ultimately provide the basis for such treatments in the future [248–250]. Furthermore, a strategy increasing the expression of specific proteasome subunits might also be useful for the protection of neurons. A novel protein has been identified in yeast that is responsible for the transcription of a number of proteasome subunits [251]. Although no mammalian homologue has yet been found, these data raise the possibility of utilizing such a protein to elevate potentially beneficial levels of proteasome expression in the CNS of human beings.
The importance of the proteasome in CNS physiology and pathology is only beginning to emerge. It is likely that future studies on this intriguing enzyme will yield further important information, particularly on the regulation of protein turnover in the CNS. Ultimately, this may lead to designing of new and useful AD therapeutics.
Neuropeptide mixture (Cerebrolysin)
Cerebrolysin (CBL) is an anti-inflammatory mixture of neuropeptides (smaller than 10kDa) obtained from porcine brain tissue [252]. Due to its complexity, this therapeutic targets different mechanisms causing neuronal death. It exhibits unique neurotrophic and neuroprotective activity and reduces amyloid burden in animal models. It also improves neurodegenerative alterations in an APP model of AD [253]. CBL regulates the activity of cyclin-dependant kinase-5 and glycogen synthases kinase 3β, which phosphorylate APP, thus reducing levels of phosphorylated APP and accumulation of APP in neuritic processes [252]. This in turn reduces the level of available APP for cleavage by the secretases. Studies in patients with mild to moderate AD have shown that CBL improves cognitive performance [254]. In addition to the neuroprotective mechanisms described above, we have recently demonstrated that CBL can also protect of neurons from complement-mediated degeneration via increasing expression of CD59 and CD55 membrane-bound CReg, while expression of CD46 remains unaffected (unpublished data). Therefore, CBL has a multilevel effect on neurons possessing anti-inflammatory, anti-apoptotic, neurotrophic and neuroprotective properties, while at the same time inducing local inhibition of complement on neuronal surfaces. Further well-designed experiments are urgently needed to further assess the beneficial properties of CBL in AD, as well as its potential in other neuropsychiatric disorders.
Conclusions
Multiple approaches to understand the pathogenesis of AD have unravelled the mechanisms of neuronal death in AD. Aβ and inflammation risk factors are considered to be the principal players in AD pathogenesis. Inflammation, Aβ and other AD-related pathological changes induce various types of toxic mechanisms that contribute to neuronal death. This indicates the complex nature of AD pathogenesis. Prevention and blockage of these mechanisms is critical for the palliative and curative therapy of AD. The advances in understanding the cellular, molecular and genetic mechanisms underlying the aetiopathogenesis of the disease lead to the identification of various potential therapeutic targets. The need for effective treatment is urgent, since current drugs for treating the cognitive impairments in AD are based on neurotransmitter replacement or modulation, which produce only mild symptomatic benefit with minimal impact on the disease process and causality. However, some very promising new strategies for fighting AD have been proposed including anti-amyloid treatment; and CBL, which protects neurons by simultaneously acting on a number of mechanisms involved in neuronal death (anti-amyloid, anti-inflammatory, anti-apoptotic, inhibition of complement system, neuroprotective, neurotrophic, etc). Due to the disease complexity, it is likely that therapeutics that aim against multiple targets will result in a more efficient management of AD and might also be effective in various forms of AD with different underlying pathophysiological mechanisms. Therefore, further extensive experimental studies, in vitro and in vivo are urgently needed.
Acknowledgments
We thank Claire Harris and B. Paul Morgan for helpful comments and discussions. R.D. was supported by the Medical Research Council, UK New Investigator Award Grant G0700102.
References
- 1.Tana S. Alzheimer’s disease – opportunities to address pharmaceutical gaps. In: Kaplan W, Laing R, editors. Priority medicines for Europe and the world – A public health approach to innovation. Geneva (Switzerland): World Health Organization; 2004. pp. 3–6. [Google Scholar]
- 2.Bertram L, McQueen M, Mullin K, et al. Systematic meta-analyses of Alzheimer disease genetic association studies: the AlzGene database. Nat Genet. 2007;39:17–23. doi: 10.1038/ng1934. [DOI] [PubMed] [Google Scholar]
- 3.Carter C. Convergence of genes implicated in Alzheimer’s disease on the cerebral cholesterol shuttle: APP, cholesterol, lipoproteins, and atherosclerosis. Neurochem Int. 2007;50:12–38. doi: 10.1016/j.neuint.2006.07.007. [DOI] [PubMed] [Google Scholar]
- 4.Selkoe D. Alzheimer’s disease results from the cerebral accumulation and cytotoxicity of amyloid beta-protein. J Alzheimers Dis. 2001;3:75–80. doi: 10.3233/jad-2001-3111. [DOI] [PubMed] [Google Scholar]
- 5.Prasad K, Cole W, Prasad K. Risk factors for Alzheimer’s disease: role of multiple antioxidants, non-steroidal anti-inflammatory and cholinergic agents alone or in combination in prevention and treatment. J Am Coll Nutr. 2002;21:506–22. doi: 10.1080/07315724.2002.10719249. [DOI] [PubMed] [Google Scholar]
- 6.Rich J, Rasmusson D, Folstein M, et al. Nonsteroidal anti-inflammatory drugs in Alzheimer’s disease. Neurology. 1995;45:51–5. doi: 10.1212/wnl.45.1.51. [DOI] [PubMed] [Google Scholar]
- 7.Akiyama H, Barger S, Barnum S, et al. Inflammation and Alzheimer’s disease. Neurobiol Aging. 2000;21:383–421. doi: 10.1016/s0197-4580(00)00124-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Bradt B, Kolb W, Cooper N. Complement-dependent proinflammatory properties of the Alzheimer’s disease beta-peptide. J Exp Med. 1998;188:431–8. doi: 10.1084/jem.188.3.431. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Breitner J, Welsh K, Helms M, et al. Delayed onset of Alzheimer’s disease with nonsteroidal anti-inflammatory and histamine H2 blocking drugs. Neurobiol Aging. 1995;16:523–30. doi: 10.1016/0197-4580(95)00049-k. [DOI] [PubMed] [Google Scholar]
- 10.Breitner J, Gau B, Welsh K, et al. Inverse association of anti-inflammatory treatments and Alzheimer’s disease: initial results of a co-twin control study. Neurology. 1994;44:227–32. doi: 10.1212/wnl.44.2.227. [DOI] [PubMed] [Google Scholar]
- 11.Andersen K, Launer L, Ott A, et al. Do nonsteroidal anti-inflammatory drugs decrease the risk for Alzheimer’s disease? The Rotterdam Study. Neurology. 1995;45:1441–5. doi: 10.1212/wnl.45.8.1441. [DOI] [PubMed] [Google Scholar]
- 12.Myllykangas-Luosujärvi R, Isomäki H. Alzheimer’s disease and rheumatoid arthritis. Br J Rheumatol. 1994;33:501–2. doi: 10.1093/rheumatology/33.5.501. [DOI] [PubMed] [Google Scholar]
- 13.McGeer P, Schulzer M, McGeer E. Arthritis and anti-inflammatory agents as possible protective factors for Alzheimer’s disease: A review of 17 epidemiologic studies. Neurology. 1996;47:425–32. doi: 10.1212/wnl.47.2.425. [DOI] [PubMed] [Google Scholar]
- 14.Rogers J, Kirby L, Hempelman S, et al. Clinical trial of indomethacin in Alzheimer’s disease. Neurology. 1993;43:1609–11. doi: 10.1212/wnl.43.8.1609. [DOI] [PubMed] [Google Scholar]
- 15.Sharif S, Hariri R, Chang V, et al. Human astrocyte production of tumour necrosis factor-alpha, interleukin-1 beta, and interleukin-6 following exposure to lipopolysaccharide endotoxin. Neurol Res. 1993;15:109–12. doi: 10.1080/01616412.1993.11740119. [DOI] [PubMed] [Google Scholar]
- 16.Shalit F, Sredni B, Stern L, et al. Elevated interleukin-6 secretion levels by mononuclear cells of Alzheimer’s patients. Neurosci Lett. 1994;174:130–2. doi: 10.1016/0304-3940(94)90003-5. [DOI] [PubMed] [Google Scholar]
- 17.Kolev M, Ruseva M, Harris C, et al. Implication of complement system and its regulators in Alzheimer’s disease. Curr Neuropharmacol. 2009;7:1–8. doi: 10.2174/157015909787602805. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Anderton B. Free radicals on the mind. Hydrogen peroxide mediates amyloid beta protein toxicity. Hum Exp Toxicol. 1994;13:716–9. [PubMed] [Google Scholar]
- 19.Chen L, Richardson J, Caldwell J, et al. Regional brain activity of free radical defense enzymes in autopsy samples from patients with Alzheimer’s disease and from nondemented controls. Int J Neurosci. 1994;75:83–90. doi: 10.3109/00207459408986291. [DOI] [PubMed] [Google Scholar]
- 20.Rozemuller J, Eikelenboom P, Pals S, et al. Microglial cells around amyloid plaques in Alzheimers disease express leukocyte adhesion molecules of the LFA-1 family. Neurosci Lett. 1989;101:288–92. doi: 10.1016/0304-3940(89)90547-8. [DOI] [PubMed] [Google Scholar]
- 21.Verbeek M, Otteholler I, Westphal J, et al. Accumulation of intercellular adhesion molecule-1 in senile plaques in brain tissue of patients with Alzheimers disease. Am J Pathol. 1994;144:104–16. [PMC free article] [PubMed] [Google Scholar]
- 22.Prasad K, Hovland A, La Rosa F, et al. Prostaglandins as putative neurotoxins in Alzheimer’s disease. Proc Soc Exp Biol Med. 1998;219:120–5. doi: 10.3181/00379727-219-44323. [DOI] [PubMed] [Google Scholar]
- 23.Lue L, Rydel R, Brigham E, et al. Inflammatory repertoire of Alzheimer’s disease and nondemented elderly microglia in vitro. Glia. 2001;35:72–9. doi: 10.1002/glia.1072. [DOI] [PubMed] [Google Scholar]
- 24.Smits H, Rijsmus A, Van Loon J, et al. Amyloid-beta-induced chemokine production in primary human macrophages and astrocytes. J Neuroimmunol. 2002;127:160–8. doi: 10.1016/s0165-5728(02)00112-1. [DOI] [PubMed] [Google Scholar]
- 25.Heneka M, Galea E, Gavriluyk V, et al. Noradrenergic depletion potentiates beta -amyloid-induced cortical inflammation: implications for Alzheimer’s disease. J Neurosci. 2002;22:2434–42. doi: 10.1523/JNEUROSCI.22-07-02434.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Joachim C, Selkoe D. The seminal role of beta-amyloid in the pathogenesis of Alzheimer disease. Alzheimer Dis Assoc Disord. 1992;6:7–34. doi: 10.1097/00002093-199205000-00003. [DOI] [PubMed] [Google Scholar]
- 27.Selkoe D. Cell biology of the amyloid beta-protein precursor and the mechanism of Alzheimer’s disease. Annu Rev Cell Biol. 1994;10:373–403. doi: 10.1146/annurev.cb.10.110194.002105. [DOI] [PubMed] [Google Scholar]
- 28.Wang G, Khatoon S, Iqbal K, et al. Brain ubiquitin is markedly elevated in Alzheimer disease. Brain Res. 1991;566:146–51. doi: 10.1016/0006-8993(91)91692-t. [DOI] [PubMed] [Google Scholar]
- 29.Kudo T, Iqbal K, Ravid R, et al. Alzheimer disease: correlation of cerebro-spinal fluid and brain ubiquitin levels. Brain Res. 1994;639:1–7. doi: 10.1016/0006-8993(94)91757-4. [DOI] [PubMed] [Google Scholar]
- 30.Checler F, Da Costa C, Ancolio K, et al. Role of the proteasome in Alzheimer’s disease. Biochim Biophys Acta. 2000;1502:133–8. doi: 10.1016/s0925-4439(00)00039-9. [DOI] [PubMed] [Google Scholar]
- 31.Rockwell P, Yuan H, Magnusson R, et al. Proteasome inhibition in neuronal cells induces a proinflammatory response manifested by upregulation of cyclooxygenase-2, its accumulation as ubiquitin conjugates, and production of the prostaglandin PGE(2) Arch Biochem Biophys. 2000;374:325–33. doi: 10.1006/abbi.1999.1646. [DOI] [PubMed] [Google Scholar]
- 32.Gregori L, Hainfeld J, Simon M, et al. Binding of amyloid beta protein to the 20 S proteasome. J Biol Chem. 1997;272:58–62. doi: 10.1074/jbc.272.1.58. [DOI] [PubMed] [Google Scholar]
- 33.Mackenzie I. Anti-inflammatory drugs and Alzheimer-type pathology in aging. Neurology. 2000;54:732–4. doi: 10.1212/wnl.54.3.732. [DOI] [PubMed] [Google Scholar]
- 34.Weggen S, Eriksen J, Das P, et al. A subset of NSAIDs lower amyloidogenic Abeta42 independently of cyclooxygenase activity. Nature. 2001;414:212–6. doi: 10.1038/35102591. [DOI] [PubMed] [Google Scholar]
- 35.Bate C, Veerhuis R, Eikelenboom P, et al. Neurones treated with cyclo-oxygenase-1 inhibitors are resistant to amyloid-beta1–42. Neuroreport. 2003;14:2099–103. doi: 10.1097/00001756-200311140-00018. [DOI] [PubMed] [Google Scholar]
- 36.Shaftel S, Griffin W, O’Banion M. The role of interleukin-1 in neuroinflammation and Alzheimer disease: an evolving perspective. J Neuroinflammation. doi: 10.1186/1742-2094-5-7. DOI: 10.1186/1742-2094-5-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Oprica M, Hjorth E, Spulber S, et al. Studies on brain volume, Alzheimer-related proteins and cytokines in mice with chronic overexpression of IL-1 receptor antagonist. J Cell Mol Med. 2007;11:810–25. doi: 10.1111/j.1582-4934.2007.00074.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Selkoe D, Schenk D. Alzheimer’s disease: Molecular understanding predicts amyloid-based therapeutics. Annu Rev Pharmacol Toxicol. 2003;43:545–84. doi: 10.1146/annurev.pharmtox.43.100901.140248. [DOI] [PubMed] [Google Scholar]
- 39.Morishima M, Ihara Y. Posttranslational modifications of tau in paired helical filaments. Dementia. 1994;5:282–8. doi: 10.1159/000106736. [DOI] [PubMed] [Google Scholar]
- 40.Caughey B, Lansbury P. Protofibrils, pores, fibrils, and neurodegeneration: Separating the responsible protein aggregates from the innocent bystanders. Annu Rev Neurosci. 2003;26:267–98. doi: 10.1146/annurev.neuro.26.010302.081142. [DOI] [PubMed] [Google Scholar]
- 41.Gschwind M, Huber G. Apoptotic cell death induced by beta-amyloid(1–42) peptide is cell type dependent. J Neurochem. 1995;65:292–300. doi: 10.1046/j.1471-4159.1995.65010292.x. [DOI] [PubMed] [Google Scholar]
- 42.Vincent B, Cisse M, Sunyach C, et al. Regulation of betaAPP and PrPc cleavage by alpha-secretase: mechanistic and therapeutic perspectives. Curr Alzheimer Res. 2008;5:202–11. doi: 10.2174/156720508783954749. [DOI] [PubMed] [Google Scholar]
- 43.Brunkan A, Goate A. Presenilin function and gamma-secretase activity. J Neurochem. 2005;93:769–92. doi: 10.1111/j.1471-4159.2005.03099.x. [DOI] [PubMed] [Google Scholar]
- 44.Wolfe M, Xia W, Ostaszewski B, et al. Two transmembrane aspartates in presenilin-1 required for presenilin endoproteolysis and gamma-secretase activity. Nature. 1999;398:513–7. doi: 10.1038/19077. [DOI] [PubMed] [Google Scholar]
- 45.De Strooper B. Aph-1, Pen-2, and Nicastrin with Presenilin generate an active gamma-Secretase complex. Neuron. 2003;38:9–12. doi: 10.1016/s0896-6273(03)00205-8. [DOI] [PubMed] [Google Scholar]
- 46.Haass C. Take five–BACE and the gamma-secretase quartet conduct Alzheimer’s amyloid beta-peptide generation. EMBO J. 2004;23:483–8. doi: 10.1038/sj.emboj.7600061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Borchelt D, Thinakaran G, Eckman C, et al. Familial Alzheimer’s disease-linked presenilin 1 variants elevate Abeta1–42/1–40 ratio in vitro and in vivo. Neuron. 1996;17:1005–13. doi: 10.1016/s0896-6273(00)80230-5. [DOI] [PubMed] [Google Scholar]
- 48.Rogaev E, Sherrington R, Rogaeva E, et al. Familial Alzheimer’s disease in kindreds with missense mutations in a gene on chromosome 1 related to the Alzheimer’s disease type 3 gene. Nature. 1995;376:775–8. doi: 10.1038/376775a0. [DOI] [PubMed] [Google Scholar]
- 49.Scheuner D, Eckman C, Jensen M, et al. Secreted amyloid beta-protein similar to that in the senile plaques of Alzheimer’s disease is increased in vivo by the presenilin 1 and 2 and APP mutations linked to familial Alzheimer’s disease. Nat Med. 1996;2:864–70. doi: 10.1038/nm0896-864. [DOI] [PubMed] [Google Scholar]
- 50.De Strooper B, Saftig P, Craessaerts K, et al. Deficiency of presenilin-1 inhibits the normal cleavage of amyloid precursor protein. Nature. 1998;391:387–90. doi: 10.1038/34910. [DOI] [PubMed] [Google Scholar]
- 51.Mastrangelo P, Mathews P, Chishti M, et al. Dissociated phenotypes in presenilin transgenic mice define functionally distinct gamma-secretases. Proc Natl Acad Sci USA. 2005;102:8972–7. doi: 10.1073/pnas.0500940102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Popescu B, Ankarcrona M. Neurons bearing presenilins: weapons for defense or suicide. J Cell Mol Med. 2000;4:249–61. doi: 10.1111/j.1582-4934.2000.tb00124.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Cowburn R, Popescu B, Ankarcrona M, et al. Presenilin-mediated signal transduction. Physiol Behav. 2007;92:93–7. doi: 10.1016/j.physbeh.2007.05.053. [DOI] [PubMed] [Google Scholar]
- 54.Canevari L, Abramov A, Duchen M. Toxicity of amyloid beta peptide: Tales of calcium, mitochondria, and oxidative stress. Neurochem Res. 2004;29:637–50. doi: 10.1023/b:nere.0000014834.06405.af. [DOI] [PubMed] [Google Scholar]
- 55.LaFerla F. Calcium dyshomeostasis and intracellular signalling in Alzheimer’s disease. Nat Rev Neurosci. 2002;3:862–72. doi: 10.1038/nrn960. [DOI] [PubMed] [Google Scholar]
- 56.Morishima Y, Gotoh Y, Zieg J, et al. beta-amyloid induces neuronal apoptosis via a mechanism that involves the c-Jun N-terminal kinase pathway and the induction of Fas ligand. J Neurosci. 2001;21:7551–60. doi: 10.1523/JNEUROSCI.21-19-07551.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Lee M, Kwon Y, Li M, et al. Neurotoxicity induces cleavage of p35 to p25 by calpain. Nature. 2000;405:360–4. doi: 10.1038/35012636. [DOI] [PubMed] [Google Scholar]
- 58.Bozyczko-Coyne D, O’Kane T, Wu Z, et al. CEP-1347/KT-7515, an inhibitor of SAPK/JNK pathway activation, promotes survival and blocks multiple events associated with A beta-induced cortical neuron apoptosis. J Neurochem. 2001;77:849–63. doi: 10.1046/j.1471-4159.2001.00294.x. [DOI] [PubMed] [Google Scholar]
- 59.Morishima-Kawashima M, Oshima N, Ogata H, et al. Effect of apolipoprotein E allele epsilon 4 on the initial phase of amyloid beta-protein accumulation in the human brain. Am J Pathol. 2000;157:2093–9. doi: 10.1016/s0002-9440(10)64847-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Mucke L, Masliah E, Yu G, et al. High-level neuronal expression of A beta(1–42) in wild-type human amyloid protein precursor transgenic mice: Synaptotoxicity without plaque formation. J Neurosci. 2000;20:4050–8. doi: 10.1523/JNEUROSCI.20-11-04050.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Lue L, Kuo Y, Roher A, et al. Soluble amyloid beta peptide concentration as a predictor of synaptic change in Alzheimer’s disease. Am J Pathol. 1999;155:853–62. doi: 10.1016/s0002-9440(10)65184-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Walsh D, Lomakin A, Benedek G, et al. Amyloid beta-protein fibrillogenesis – Detection of a protofibrillar intermediate. J Biol Chem. 1997;272:22364–72. doi: 10.1074/jbc.272.35.22364. [DOI] [PubMed] [Google Scholar]
- 63.Lambert M, Barlow A, Chromy B, et al. Diffusible, nonfibrillar ligands derived from Abeta1–42 are potent central nervous system neurotoxins. Proc Natl Acad Sci USA. 1998;95:6448–53. doi: 10.1073/pnas.95.11.6448. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Wang H, Pasternak J, Kuo H, et al. Soluble oligomers of beta amyloid (1–42) inhibit long-term potentiation but not long-term depression in rat dentate gyrus. Brain Res. 2002;924:133–40. doi: 10.1016/s0006-8993(01)03058-x. [DOI] [PubMed] [Google Scholar]
- 65.Klein W. Abeta toxicity in Alzheimer’s disease: globular oligomers (ADDLs) as new vaccine and drug targets. Neurochem Int. 2002;41:345–52. doi: 10.1016/s0197-0186(02)00050-5. [DOI] [PubMed] [Google Scholar]
- 66.Shankar G, Li S, Mehta T, et al. Amyloid-beta protein dimers isolated directly from Alzheimer’s brains impair synaptic plasticity and memory. Nat Med. 2008;14:837–42. doi: 10.1038/nm1782. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Naylor R, Hill A, Barnham K. Is covalently crosslinked Abeta responsible for synaptotoxicity in Alzheimer’s disease. Curr Alzheimer Res. 2008;5:533–9. doi: 10.2174/156720508786898433. [DOI] [PubMed] [Google Scholar]
- 68.Asghar S, Pasch M. Therapeutic inhibition of the complement system. Y2K update. Front Biosci. 2000;5:E63–81. doi: 10.2741/asghar. [DOI] [PubMed] [Google Scholar]
- 69.Silva C, Rio M, Cruz C. Local immunoglobulin synthesis and blood-brain barrier assessment in subacute sclerosing panencephalitis. Eur Neurol. 1985;24:128–33. doi: 10.1159/000115773. [DOI] [PubMed] [Google Scholar]
- 70.Barnum S. Complement biosynthesis in the central nervous system. Crit Rev Oral Biol Med. 1995;6:132–46. doi: 10.1177/10454411950060020301. [DOI] [PubMed] [Google Scholar]
- 71.Emmerling MR, Watson MD, Raby CA, et al. The role of complement in Alzheimer’s disease pathology. Bichemica et Biophysica Acta. 2000;1502:158–71. doi: 10.1016/s0925-4439(00)00042-9. [DOI] [PubMed] [Google Scholar]
- 72.Gasque P, Fontaine M, Morgan B. Complement expression in human brain – biosynthesis of terminal pathway components and regulators in human glial cells and cell lines. J Immunol. 1995;154:4726–33. [PubMed] [Google Scholar]
- 73.Vedeler C, Ulvestad E, Bjorge L, et al. The expression of CD59 in normal human nervos tissue. Immunol. 1994;82:542–7. [PMC free article] [PubMed] [Google Scholar]
- 74.Pasinetti G, Johnson S, Oda T, et al. Clusterin (SGP-2) – a multifunctional glycoprotein with regional expression in astrocytes and neurons of the adult rat brain. J Comp Neurol. 1994;339:387–400. doi: 10.1002/cne.903390307. [DOI] [PubMed] [Google Scholar]
- 75.Yang L, Li R, Meri S, Rogers J, et al. Deficiency of complement defense protein CD59 may contribute to neurodegeneration in Alzheimer’s disease. J Neurosci. 2000;20:7505–9. doi: 10.1523/JNEUROSCI.20-20-07505.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Osaka H, Mukherjee P, Aisen P, et al. Complement-derived anaphylatoxin C5a protects against glutamate-mediated neurotoxicity. J Cell Biochem. 1999;73:303–11. [PubMed] [Google Scholar]
- 77.Pasinetti G. Inflammatory mechanisms in neurodegeneration and Alzheimer’s disease: the role of the complement system. Neurobiol Aging. 1996;17:707–16. doi: 10.1016/0197-4580(96)00113-3. [DOI] [PubMed] [Google Scholar]
- 78.Wyss-Coray T, Yan F, Lin A, et al. Prominent neurodegeneration and increased plaque formation in complement-inhibited Alzheimer’s mice. Proc Natl Acad Sci USA. 2002;99:10837–42. doi: 10.1073/pnas.162350199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Pisalyaput K, Tenner A. Complement component C1q inhibits beta-amyloid- and serum amyloid P-induced neurotoxicity via caspase- and calpain-independent mechanisms. J Neurochem. 2008;104:696–707. doi: 10.1111/j.1471-4159.2007.05012.x. [DOI] [PubMed] [Google Scholar]
- 80.Maier M, Peng Y, Jiang L, et al. Complement C3 deficiency leads to accelerated amyloid beta plaque deposition and neurodegeneration and modulation of the microglia/macrophage phenotype in amyloid precursor protein transgenic mice. J Neurosci. 2008;28:6333–41. doi: 10.1523/JNEUROSCI.0829-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Gasque P, Thomas A, Fontaine M, et al. Complement activation on human neuroblastoma cell lines in vitro: route of activation and expression of functional complement regulatory proteins. J Neuroimmunol. 1996;66:29–40. doi: 10.1016/0165-5728(96)00015-x. [DOI] [PubMed] [Google Scholar]
- 82.Sano M, Ernesto C, Thomas R, et al. A controlled trial of selegiline, alpha-tocopherol, or both as treatment for Alzheimer’s disease. The Alzheimer’s Disease Cooperative Study. N Engl J Med. 1997;336:1216–22. doi: 10.1056/NEJM199704243361704. [DOI] [PubMed] [Google Scholar]
- 83.Birkmayer J. Coenzyme nicotinamide adenine dinucleotide: new therapeutic approach for improving dementia of the Alzheimer type. Ann Clin Lab Sci. 1996;26:1–9. [PubMed] [Google Scholar]
- 84.Misonou H, Morishima-Kawashima M, et al. Oxidative stress induces intracellular accumulation of amyloid beta-protein (A beta) in human neuroblastoma cells. Biochemistry. 2000;39:6951–9. doi: 10.1021/bi000169p. [DOI] [PubMed] [Google Scholar]
- 85.Koppaka V, Axelsen P. Accelerated accumulation of amyloid beta proteins on oxidatively damaged lipid membranes. Biochemistry. 2000;39:10011–6. doi: 10.1021/bi000619d. [DOI] [PubMed] [Google Scholar]
- 86.Schubert D, Behl C, Lesley R, et al. Amyloid peptides are toxic via a common oxidative mechanism. Proc Natl Acad Sci USA. 1995;92:1989–93. doi: 10.1073/pnas.92.6.1989. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Behl C, Davis J, Cole G, et al. Vitamin E protects nerve cells from amyloid beta protein toxicity. Biochem Biophys Res Commun. 1992;186:944–50. doi: 10.1016/0006-291x(92)90837-b. [DOI] [PubMed] [Google Scholar]
- 88.Zaman Z, Roche S, Fielden P, et al. Plasma concentrations of vitamin E and carotenoids in Alzheimers disease. Age Ageing. 1992;21:91–4. doi: 10.1093/ageing/21.2.91. [DOI] [PubMed] [Google Scholar]
- 89.Sims N, Bowen D, Neary D, et al. Metabolic processes in Alzheimers disease adenine nucleotide content and production of (CO2)-C-14 from [U-C-14] glucose in vitro in human neocortex. J Neurochem. 1983;41:1329–34. doi: 10.1111/j.1471-4159.1983.tb00829.x. [DOI] [PubMed] [Google Scholar]
- 90.Saito K, Elce J, Hamos J, et al. Widespread activation of calcium-activated neutral proteinase (calpain) in the brain in Alzheimer disease – a potential molecular basis for neuronal degeneration. Proc Natl Acad Sci USA. 1993;90:2628–32. doi: 10.1073/pnas.90.7.2628. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Zhou Y, Richardson J, Mombourquette M, et al. Free radical formation in autopsy samples of Alzheimer and control cortex. Neurosci Lett. 1995;195:89–92. doi: 10.1016/0304-3940(94)11787-j. [DOI] [PubMed] [Google Scholar]
- 92.Simic G, Lucassen P, Krsnik Z, et al. nNOS expression in reactive astrocytes correlates with increased cell death related DNA damage in the hippocampus and entorhinal cortex in Alzheimer’s disease. Exp Neurol. 2000;165:12–26. doi: 10.1006/exnr.2000.7448. [DOI] [PubMed] [Google Scholar]
- 93.Conrad C, Marshall P, Talent J, et al. Oxidized proteins in Alzheimer’s plasma. Biochem Biophys Res Commun. 2000;275:678–81. doi: 10.1006/bbrc.2000.3356. [DOI] [PubMed] [Google Scholar]
- 94.Keller J, Markesbery W. Proteasome inhibition results in increased poly-ADP-ribosylation: Implications for neuron death. J Neurosci Res. 2000;61:436–42. doi: 10.1002/1097-4547(20000815)61:4<436::AID-JNR10>3.0.CO;2-Z. [DOI] [PubMed] [Google Scholar]
- 95.Qiu J, Asai A, Chi S, et al. Proteasome inhibitors induce cytochrome c-caspase-3-like protease-mediated apoptosis in cultured cortical neurons. J Neurosci. 2000;20:259–65. doi: 10.1523/JNEUROSCI.20-01-00259.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Pasquini L, Moreno M, Adamo A, et al. Lactacystin, a specific inhibitor of the proteasome, induces apoptosis and activates caspase-3 in cultured cerebellar granule cells. J Neurosci Res. 2000;59:601–11. doi: 10.1002/(SICI)1097-4547(20000301)59:5<601::AID-JNR3>3.0.CO;2-1. [DOI] [PubMed] [Google Scholar]
- 97.Wagenknecht B, Hermisson M, Groscurth P, et al. Proteasome inhibitor-induced apoptosis of glioma cells involves the processing of multiple caspases and cytochrome c release. J Neurochem. 2000;75:2288–97. doi: 10.1046/j.1471-4159.2000.0752288.x. [DOI] [PubMed] [Google Scholar]
- 98.Taglialatela G, Kaufmann J, Trevino A, et al. Central nervous system DNA fragmentation induced by the inhibition of nuclear factor kappa B. Neuroreport. 1998;9:489–93. doi: 10.1097/00001756-199802160-00024. [DOI] [PubMed] [Google Scholar]
- 99.Kitagawa H, Tani E, Ikemoto H, et al. Proteasome inhibitors induce mitochondria-independent apoptosis in human glioma cells. FEBS Lett. 1999;443:181–6. doi: 10.1016/s0014-5793(98)01709-8. [DOI] [PubMed] [Google Scholar]
- 100.Marambaud P, Chevallier N, Barelli H, et al. Proteasome contributes to the alpha-secretase pathway of amyloid precursor protein in human cells. J Neurochem. 1997;68:698–703. doi: 10.1046/j.1471-4159.1997.68020698.x. [DOI] [PubMed] [Google Scholar]
- 101.Skovronsky D, Pijak D, Doms R, et al. A distinct ER/IC gamma-secretase competes with the proteasome for cleavage of APP. Biochemistry. 2000;39:810–7. doi: 10.1021/bi991728z. [DOI] [PubMed] [Google Scholar]
- 102.Honda T, Yasutake K, Nihonmatsu N, et al. Dual roles of proteasome in the metabolism of presenilin 1. J Neurochem. 1999;72:255–61. doi: 10.1046/j.1471-4159.1999.0720255.x. [DOI] [PubMed] [Google Scholar]
- 103.Ingano L, Lentini K, Kovacs I, et al. Cytoplasmic presenilin aggregates in proteasome inhibitor-treated cells. Mol Basis Dementia. 2000;920:259–60. doi: 10.1111/j.1749-6632.2000.tb06933.x. [DOI] [PubMed] [Google Scholar]
- 104.Kwak S, Masaki T, Ishiura S, et al. Multicatalytic proteinase is present in Lewy bodies and neurofibrillary tangles in diffuse Lewy body disease brains. Neurosci Lett. 1991;128:21–4. doi: 10.1016/0304-3940(91)90751-e. [DOI] [PubMed] [Google Scholar]
- 105.Masaki T, Ishiura S, Sugita H, et al. Multicatalytic proteinsase is associated with characteristic oval structures in cortical Lewy bodies – an immunocytochemical study with light and electron microscopy. J Neurol Sci. 1994;122:127–34. doi: 10.1016/0022-510x(94)90288-7. [DOI] [PubMed] [Google Scholar]
- 106.Canu N, Barbato C, Ciotti M, et al. Proteasome involvement and accumulation of ubiquitinated proteins in cerebellar granule neurons undergoing apoptosis. J Neurosci. 2000;20:589–99. doi: 10.1523/JNEUROSCI.20-02-00589.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Favit A, Grimaldi M, Alkon D. Prevention of beta-amyloid neurotoxicity by blockade of the ubiquitin-proteasome proteolytic pathway. J Neurochem. 2000;75:1258–63. doi: 10.1046/j.1471-4159.2000.0751258.x. [DOI] [PubMed] [Google Scholar]
- 108.Grilli M, Memo M. Possible role of NF-kappa B and p53 in the glutamate-induced pro-apoptotic neuronal pathway. Cell Death Differ. 1999;6:22–7. doi: 10.1038/sj.cdd.4400463. [DOI] [PubMed] [Google Scholar]
- 109.Mattson M, Culmsee C, Yu Z, et al. Roles of nuclear factor kappa B in neuronal survival and plasticity. J Neurochem. 2000;74:443–56. doi: 10.1046/j.1471-4159.2000.740443.x. [DOI] [PubMed] [Google Scholar]
- 110.Simons M, Keller P, De Strooper B, et al. Cholesterol depletion inhibits the generation of beta-amyloid in hippocampal neurons. Proc Natl Acad Sci USA. 1998;95:6460–4. doi: 10.1073/pnas.95.11.6460. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Refolo L, Pappolla M, Malester B, et al. Hypercholesterolemia accelerates the Alzheimer’s amyloid pathology in a transgenic mouse model. Neurobiol Disease. 2000;7:321–31. doi: 10.1006/nbdi.2000.0304. [DOI] [PubMed] [Google Scholar]
- 112.Refolo L, Pappolla M, LaFrancois J, et al. A cholesterol-lowering drug reduces beta-amyloid pathology in a transgenic mouse model of Alzheimer’s disease. Neurobiol Disease. 2001;8:890–9. doi: 10.1006/nbdi.2001.0422. [DOI] [PubMed] [Google Scholar]
- 113.Sparks D, Scheff S, Hunsaker J, et al. Induction of Alzheimer-like β-amyloid immunoreactivity in the brains of rabbits with dietary cholesterol. Exp Neurol. 1994;126:88–94. doi: 10.1006/exnr.1994.1044. [DOI] [PubMed] [Google Scholar]
- 114.Sparks D. Intraneuronal beta-amyloid immunoreactivity in the CNS. Neurobiol Aging. 1996;17:291–9. doi: 10.1016/0197-4580(95)02067-5. [DOI] [PubMed] [Google Scholar]
- 115.Esiri M, Biddolph S, Morris C. Prevalence of Alzheimer plaques in AIDS. J Neurol Neurosurg Psychiatry. 1998;65:29–33. doi: 10.1136/jnnp.65.1.29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Kountouras J, Tsolaki M, Gavalas E, et al. Relationship between Helicobacter pylori infection and Alzheimer disease. Neurology. 2006;66:938–40. doi: 10.1212/01.wnl.0000203644.68059.5f. [DOI] [PubMed] [Google Scholar]
- 117.Itzhaki R, Wozniak M, Appelt D, et al. Infiltration of the brain by pathogens causes Alzheimer’s disease. Neurobiol Aging. 2004;25:619–27. doi: 10.1016/j.neurobiolaging.2003.12.021. [DOI] [PubMed] [Google Scholar]
- 118.Ball M. The essential lesion of Alzheimer disease: a surprise in retrospect. J Alzheimers Dis. 2006;9:29–33. doi: 10.3233/jad-2006-9s304. [DOI] [PubMed] [Google Scholar]
- 119.Itzhaki R, Wozniak M. Herpes simplex virus type 1, apolipoprotein E, and cholesterol: a dangerous liaison in Alzheimer’s disease and other disorders. Prog Lipid Res. 2006;45:73–90. doi: 10.1016/j.plipres.2005.11.003. [DOI] [PubMed] [Google Scholar]
- 120.Shipley S, Parkin E, Itzhaki R, et al. Herpes simplex virus interferes with amyloid precursor protein processing. BMC Microbiol. doi: 10.1186/1471-2180-5-48. DOI: 10.1186/1471-2180-5-48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Satpute-Krishnan P, DeGiorgis J, et al. Fast anterograde transport of herpes simplex virus: role for the amyloid precursor protein of Alzheimer’s disease. Aging Cell. 2003;2:305–18. doi: 10.1046/j.1474-9728.2003.00069.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Wozniak M, Itzhaki R, Shipley S, et al. Herpes simplex virus infection causes cellular beta-amyloid accumulation and secretase upregulation. Neurosci Lett. 2007;429:95–100. doi: 10.1016/j.neulet.2007.09.077. [DOI] [PubMed] [Google Scholar]
- 123.Heyman A, Wilkinson W, Stafford J, et al. Alzheimer’s disease: a study of epidemiological aspects. Ann Neurol. 1984;15:335–41. doi: 10.1002/ana.410150406. [DOI] [PubMed] [Google Scholar]
- 124.French L, Schuman L, Mortimer J, et al. A case-control study of dementia of the Alzheimer type. Am J Epidemiol. 1985;121:414–21. doi: 10.1093/oxfordjournals.aje.a114013. [DOI] [PubMed] [Google Scholar]
- 125.Mortimer J, Van Duijn C, Chandra V, et al. Head trauma as a risk factor for Alzheimer’s disease: a collaborative re-analysis of case-control studies. Int J Epidemiol. 1991;20:S28–35. doi: 10.1093/ije/20.supplement_2.s28. EURODEM Risk Factors Research Group. [DOI] [PubMed] [Google Scholar]
- 126.Schofield P, Tang M, Marder K, et al. Alzheimer’s disease after remote head injury: an incidence study. J Neurol Neurosurg Psychiatry. 1997;62:119–24. doi: 10.1136/jnnp.62.2.119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Yamada M, Sasaki H, Mimori Y, et al. Prevalence and risks of dementia in the Japanese population: RERF’s adult health study Hiroshima subjects. Radiation Effects Research Foundation. J Am Geriatr Soc. 1999;47:189–95. doi: 10.1111/j.1532-5415.1999.tb04577.x. [DOI] [PubMed] [Google Scholar]
- 128.Jamieson G, Maitland N, Wilcock G, et al. Latent herpes simplex virus type 1 in normal and Alzheimer’s disease brains. J Med Virol. 1991;33:224–7. doi: 10.1002/jmv.1890330403. [DOI] [PubMed] [Google Scholar]
- 129.Lin W, Shang D, Itzhaki R. Neurotropic viruses and Alzheimer disease. Interaction of herpes simplex type 1 virus and apolipoprotein E in the etiology of the disease. Mol Chem Neuropathol. 1996;28:135–41. doi: 10.1007/BF02815215. [DOI] [PubMed] [Google Scholar]
- 130.Langenberg A, Corey L, Ashley R, et al. A prospective study of new infections with herpes simplex virus type 1 and type 2. Chiron HSV Vaccine Study Group. N Engl J Med. 1999;341:1432–8. doi: 10.1056/NEJM199911043411904. [DOI] [PubMed] [Google Scholar]
- 131.Hill J, Steiner I, Matthews K, et al. Statins lower the risk of developing Alzheimer’s disease by limiting lipid raft endocytosis and decreasing the neuronal spread of Herpes simplex virus type 1. Med Hypotheses. 2005;64:53–8. doi: 10.1016/j.mehy.2003.12.058. [DOI] [PubMed] [Google Scholar]
- 132.Petryshyn R, Chen J, Danley L, et al. Effect of interferon on protein translation during growth stages of 3T3 cells. Arch Biochem Biophys. 1996;326:290–7. doi: 10.1006/abbi.1996.0078. [DOI] [PubMed] [Google Scholar]
- 133.Hiremath M, Mikhael A, Taylor L, et al. Complex regulation of the Csk homologous kinase (Chk) by IL-4 family cytokines and IFN-gamma in human peripheral blood monocytes. Mol Immunol. 2004;41:901–10. doi: 10.1016/j.molimm.2004.04.025. [DOI] [PubMed] [Google Scholar]
- 134.Donev RM, Cole DS, Sivasankar B, et al. p53 regulates cellular resistance to complement lysis through enhanced expression of CD59. Cancer Res. 2006;66:2451–8. doi: 10.1158/0008-5472.CAN-05-3191. [DOI] [PubMed] [Google Scholar]
- 135.Donev R, Newall A, Thome J, et al. A role for SC35 and hnRNPA1 in the determination of amyloid precursor protein isoforms. Mol Psychiatry. 2007;12:681–90. doi: 10.1038/sj.mp.4001971. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Yamamoto M, Kiyota T, Horiba M, et al. Interferon-gamma and tumor necrosis factor-alpha regulate amyloid-beta plaque deposition and beta-secretase expression in Swedish mutant APP transgenic mice. Am J Pathol. 2007;170:680–92. doi: 10.2353/ajpath.2007.060378. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Hardwicke M, Sandri-Goldin R. The herpes simplex virus regulatory protein ICP27 contributes to the decrease in cellular mRNA levels during infection. J Virol. 1994;68:4797–810. doi: 10.1128/jvi.68.8.4797-4810.1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Bryant H, Wadd S, Lamond A, et al. Herpes simplex virus IE63 (ICP27) protein interacts with spliceosome-associated protein 145 and inhibits splicing prior to the first catalytic step. J Virol. 2001;75:4376–85. doi: 10.1128/JVI.75.9.4376-4385.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Lindberg A, Kreivi J. Splicing inhibition at the level of spliceosome assembly in the presence of herpes simplex virus protein ICP27. Virology. 2002;294:189–98. doi: 10.1006/viro.2001.1301. [DOI] [PubMed] [Google Scholar]
- 140.Sciabica K, Dai Q, Sandri-Goldin R. ICP27 interacts with SRPK1 to mediate HSV splicing inhibition by altering SR protein phosphorylation. EMBO J. 2003;22:1608–19. doi: 10.1093/emboj/cdg166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Klajnert B, Cortijo-Arellano M, Cladera J, et al. Influence of dendrimer’s structure on its activity against amyloid fibril formation. Biochem Biophys Res Commun. 2006;345:21–8. doi: 10.1016/j.bbrc.2006.04.041. [DOI] [PubMed] [Google Scholar]
- 142.Yanagisawa K. Cholesterol and amyloid beta fibrillogenesis. Subcell Biochem. 2005;38:179–202. doi: 10.1007/0-387-23226-5_9. [DOI] [PubMed] [Google Scholar]
- 143.Cummings J. Alzheimer’s disease. N Engl J Med. 2004;351:56–67. doi: 10.1056/NEJMra040223. [DOI] [PubMed] [Google Scholar]
- 144.Francis P, Palmer A, Snape M, et al. The cholinergic hypothesis of Alzheimer’s disease: a review of progress. J Neurol Neurosurg Psychiatry. 1999;66:137–47. doi: 10.1136/jnnp.66.2.137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Cummings J, Kaufer D. Neuropsychiatric aspects of Alzheimer’s disease: the cholinergic hypothesis revisited. Neurology. 1996;47:876–83. doi: 10.1212/wnl.47.4.876. [DOI] [PubMed] [Google Scholar]
- 146.Perry E, Gibson P, Blessed G, et al. Neurotransmitter enzyme abnormalities in senile dementia. Choline acetyltransferase and glutamic acid decarboxylase activities in necropsy brain tissue. J Neurol Sci. 1977;34:247–65. doi: 10.1016/0022-510x(77)90073-9. [DOI] [PubMed] [Google Scholar]
- 147.Wilcock G, Esiri M, Bowen D, et al. Alzheimer’s disease. Correlation of cortical choline acetyltransferase activity with the severity of dementia and histological abnormalities. J Neurol Sci. 1982;57:407–17. doi: 10.1016/0022-510x(82)90045-4. [DOI] [PubMed] [Google Scholar]
- 148.Perry E, Tomlinson B, Blessed G, et al. Correlation of cholinergic abnormalities with senile plaques and mental test scores in senile dementia. Br Med J. 1978;2:1457–9. doi: 10.1136/bmj.2.6150.1457. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Wilkinson D, Francis P, Schwam E, et al. Cholinesterase inhibitors used in the treatment of Alzheimer’s disease: the relationship between pharmacological effects and clinical efficacy. Drugs Aging. 2004;21:453–78. doi: 10.2165/00002512-200421070-00004. [DOI] [PubMed] [Google Scholar]
- 150.Katsuki H, Izumi Y, Zorumski C. Removal of extracellular calcium after conditioning stimulation disrupts long-term potentiation in the CA1 region of rat hippocampal slices. Neuroscience. 1997;76:1113–9. doi: 10.1016/s0306-4522(97)80003-6. [DOI] [PubMed] [Google Scholar]
- 151.Lipton S. Failures and successes of NMDA receptor antagonists: molecular basis for the use of open-channel blockers like memantine in the treatment of acute and chronic neurologic insults. NeuroRx. 2004;1:101–10. doi: 10.1602/neurorx.1.1.101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Reisberg B, Doody R, Stöffler A, et al. Memantine in moderate-to-severe Alzheimer’s disease. N Engl J Med. 2003;348:1333–41. doi: 10.1056/NEJMoa013128. [DOI] [PubMed] [Google Scholar]
- 153.Winblad B, Poritis N. Memantine in severe dementia: results of the 9M-Best Study (Benefit and efficacy in severely demented patients during treatment with memantine) Int J Geriatr Psychiatry. 1999;14:135–46. doi: 10.1002/(sici)1099-1166(199902)14:2<135::aid-gps906>3.0.co;2-0. [DOI] [PubMed] [Google Scholar]
- 154.Roberds S, Anderson J, Basi G, et al. BACE knockout mice are healthy despite lacking the primary beta-secretase activity in brain: implications for Alzheimer’s disease therapeutics. Hum Mol Genet. 2001;10:1317–24. doi: 10.1093/hmg/10.12.1317. [DOI] [PubMed] [Google Scholar]
- 155.Fukumoto H, Rosene D, Moss M, et al. Beta-secretase activity increases with aging in human, monkey, and mouse brain. Am J Pathol. 2004;164:719–25. doi: 10.1016/s0002-9440(10)63159-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Leung D, Abbenante G, Fairlie D. Protease inhibitors: current status and future prospects. J Med Chem. 2000;43:305–41. doi: 10.1021/jm990412m. [DOI] [PubMed] [Google Scholar]
- 157.Hong L, Koelsch G, Lin X, et al. Structure of the protease domain of memapsin 2 (beta-secretase) complexed with inhibitor. Science. 2000;290:150–3. doi: 10.1126/science.290.5489.150. [DOI] [PubMed] [Google Scholar]
- 158.Asai M, Hattori C, Iwata N, et al. The novel beta-secretase inhibitor KMI-429 reduces amyloid beta peptide production in amyloid precursor protein transgenic and wild-type mice. J Neurochem. 2006;96:533–40. doi: 10.1111/j.1471-4159.2005.03576.x. [DOI] [PubMed] [Google Scholar]
- 159.Ghosh A, Kumaragurubaran N, Hong L, et al. Potent memapsin 2 (beta-secretase) inhibitors: design, synthesis, protein-ligand X-ray structure, and in vivo evaluation. Bioorg Med Chem Lett. 2008;18:1031–6. doi: 10.1016/j.bmcl.2007.12.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Hamada Y, Ohta H, Miyamoto N, et al. Novel non-peptidic and small-sized BACE1 inhibitors. Bioorg Med Chem Lett. 2008;18:1643–7. doi: 10.1016/j.bmcl.2008.01.056. [DOI] [PubMed] [Google Scholar]
- 161.Hussain I, Hawkins J, Harrison D, et al. Oral administration of a potent and selective non-peptidic BACE-1 inhibitor decreases beta-cleavage of amyloid precursor protein and amyloid-beta production in vivo. J Neurochem. 2007;100:802–9. doi: 10.1111/j.1471-4159.2006.04260.x. [DOI] [PubMed] [Google Scholar]
- 162.Wong P, Zheng H, Chen H, et al. Presenilin 1 is required for Notch1 and DII1 expression in the paraxial mesoderm. Nature. 1997;387:288–92. doi: 10.1038/387288a0. [DOI] [PubMed] [Google Scholar]
- 163.Shen J, Bronson R, Chen D, et al. Skeletal and CNS defects in presenilin-1-deficient mice. Cell. 1997;89:629–39. doi: 10.1016/s0092-8674(00)80244-5. [DOI] [PubMed] [Google Scholar]
- 164.Selkoe D, Kopan R. Notch and Presenilin: regulated intramembrane proteolysis links development and degeneration. Annu Rev Neurosci. 2003;26:565–97. doi: 10.1146/annurev.neuro.26.041002.131334. [DOI] [PubMed] [Google Scholar]
- 165.Searfoss G, Jordan W, Calligaro D, et al. Adipsin, a biomarker of gastrointestinal toxicity mediated by a functional gamma-secretase inhibitor. J Biol Chem. 2003;278:46107–16. doi: 10.1074/jbc.M307757200. [DOI] [PubMed] [Google Scholar]
- 166.Wong G, Manfra D, Poulet F, et al. Chronic treatment with the gamma-secretase inhibitor LY-411,575 inhibits beta-amyloid peptide production and alters lymphopoiesis and intestinal cell differentiation. J Biol Chem. 2004;279:12876–82. doi: 10.1074/jbc.M311652200. [DOI] [PubMed] [Google Scholar]
- 167.Siemers E, Quinn J, Kaye J, et al. Effects of a gamma-secretase inhibitor in a randomized study of patients with Alzheimer disease. Neurology. 2006;66:602–4. doi: 10.1212/01.WNL.0000198762.41312.E1. [DOI] [PubMed] [Google Scholar]
- 168.Barten D, Guss V, Corsa J, et al. Dynamics of {beta}-amyloid reductions in brain, cerebrospinal fluid, and plasma of {beta}-amyloid precursor protein transgenic mice treated with a {gamma}-secretase inhibitor. J Pharmacol Exp Ther. 2005;312:635–43. doi: 10.1124/jpet.104.075408. [DOI] [PubMed] [Google Scholar]
- 169.Anderson J, Holtz G, Baskin P, et al. Reductions in beta-amyloid concentrations in vivo by the gamma-secretase inhibitors BMS-289948 and BMS-299897. Biochem Pharmacol. 2005;69:689–98. doi: 10.1016/j.bcp.2004.11.015. [DOI] [PubMed] [Google Scholar]
- 170.Kopan R, Ilagan M. Gamma-secretase: proteasome of the membrane. Nat Rev Mol Cell Biol. 2004;5:499–504. doi: 10.1038/nrm1406. [DOI] [PubMed] [Google Scholar]
- 171.Wolfe M. Inhibition and modulation of gamma-secretase for Alzheimer’s disease. Neurotherapeutics. 2008;5:391–8. doi: 10.1016/j.nurt.2008.05.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Tomita T. Secretase inhibitors and modulators for Alzheimer’s disease treatment. Expert Rev Neurother. 2009;9:661–79. doi: 10.1586/ern.09.24. [DOI] [PubMed] [Google Scholar]
- 173.MRC/BHF. Heart Protection Study of antioxidant vitamin supplementation in 20,536 high-risk individuals: a randomised placebo-controlled trial. Lancet. 2002;360:23–33. doi: 10.1016/S0140-6736(02)09328-5. [DOI] [PubMed] [Google Scholar]
- 174.Luchsinger J, Tang M, Shea S, et al. Antioxidant vitamin intake and risk of Alzheimer disease. Arch Neurol. 2003;60:203–8. doi: 10.1001/archneur.60.2.203. [DOI] [PubMed] [Google Scholar]
- 175.Laurin D, Masaki K, Foley D, et al. Midlife dietary intake of antioxidants and risk of late-life incident dementia: the Honolulu-Asia Aging Study. Am J Epidemiol. 2004;159:959–67. doi: 10.1093/aje/kwh124. [DOI] [PubMed] [Google Scholar]
- 176.Miller Er, Pastor-Barriuso R, Dalal D. Meta-analysis: high-dosage vitamin E supplementation may increase all-cause mortality. Ann Intern Med. 2005;142:37–46. doi: 10.7326/0003-4819-142-1-200501040-00110. [DOI] [PubMed] [Google Scholar]
- 177.Cummings J. The role of cholinergic agents in the management of behavioural disturbances in Alzheimer’s disease. Int J Neuropsychopharmacol. 2000;3:21–9. doi: 10.1017/S1461145700001917. [DOI] [PubMed] [Google Scholar]
- 178.Solomon P, Adams F, Silver A, et al. Ginkgo for memory enhancement: a randomized controlled trial. JAMA. 2002;288:835–40. doi: 10.1001/jama.288.7.835. [DOI] [PubMed] [Google Scholar]
- 179.Etminan M, Gill S, Samii A. Effect of non-steroidal anti-inflammatory drugs on risk of Alzheimer’s disease: systematic review and meta-analysis of observational studies. BMJ. 2003;327:128. doi: 10.1136/bmj.327.7407.128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.In T’ Veld B, Ruitenberg A, Hofman A, et al. Nonsteroidal antiinflammatory drugs and the risk of Alzheimer’s disease. N Engl J Med. 2001;345:1515–21. doi: 10.1056/NEJMoa010178. [DOI] [PubMed] [Google Scholar]
- 181.Zandi P, Anthony J, Hayden K, et al. Reduced incidence of AD with NSAID but not H2 receptor antagonists: the Cache County Study. Neurology. 2002;59:880–6. doi: 10.1212/wnl.59.6.880. [DOI] [PubMed] [Google Scholar]
- 182.Beard C, Waring S, O’Brien P, et al. Nonsteroidal anti-inflammatory drug use and Alzheimer’s disease: a case-control study in Rochester, Minnesota, 1980 through 1984. Mayo Clin Proc. 1998;73:951–5. doi: 10.4065/73.10.951. [DOI] [PubMed] [Google Scholar]
- 183.Aisen P, Schafer K, Grundman M, et al. Effects of rofecoxib or naproxen vs placebo on Alzheimer disease progression: a randomized controlled trial. JAMA. 2003;289:2819–26. doi: 10.1001/jama.289.21.2819. [DOI] [PubMed] [Google Scholar]
- 184.Van Gool W, Weinstein H, Scheltens P, et al. Effect of hydroxychloroquine on progression of dementia in early Alzheimer’s disease: an 18-month randomised, double-blind, placebo-controlled study. Lancet. 2001;358:455–60. doi: 10.1016/s0140-6736(01)05623-9. [DOI] [PubMed] [Google Scholar]
- 185.Aisen P. Anti-inflammatory therapy for Alzheimer’s disease: implications of the prednisone trial. Acta Neurol Scand Suppl. 2000;176:85–9. doi: 10.1034/j.1600-0404.2000.00312.x. [DOI] [PubMed] [Google Scholar]
- 186.Aisen P, Schmeidler J, Pasinetti G. Randomized pilot study of nimesulide treatment in Alzheimer’s disease. Neurology. 2002;58:1050–4. doi: 10.1212/wnl.58.7.1050. [DOI] [PubMed] [Google Scholar]
- 187.Scharf S, Mander A, Ugoni A, et al. A double-blind, placebo-controlled trial of diclofenac/misoprostol in Alzheimer’s disease. Neurology. 1999;53:197–201. doi: 10.1212/wnl.53.1.197. [DOI] [PubMed] [Google Scholar]
- 188.Thomas T, Rhodin J. Vascular actions of estrogen and Alzheimer’s disease. Ann N Y Acad Sci. 2000;903:501–9. doi: 10.1111/j.1749-6632.2000.tb06406.x. [DOI] [PubMed] [Google Scholar]
- 189.Koh K, Cardillo C, Bui M, et al. Vascular effects of estrogen and cholesterol-lowering therapies in hypercholesterolemic postmenopausal women. Circulation. 1999;99:354–60. doi: 10.1161/01.cir.99.3.354. [DOI] [PubMed] [Google Scholar]
- 190.Gewurz H, Clark D, Cooper M, et al. Effect of cobra venom-induced inhibition of complement activity on allograft and xenograft rejection reactions. Transplantation. 1967;5:1296–303. doi: 10.1097/00007890-196709000-00008. [DOI] [PubMed] [Google Scholar]
- 191.Maillard J, Zarco R. [Decomplementization by a factor extracted from cobra venom. Effect on several immune reactions of the guinea pig and rat] Ann Inst Pasteur. 1968;114:756–74. [PubMed] [Google Scholar]
- 192.Cochrane C, Müller-Eberhard H, Aikin B. Depletion of plasma complement in vivo by a protein of cobra venom: its effect on various immunologic reactions. J Immunol. 1970;105:55–69. [PubMed] [Google Scholar]
- 193.Phillips G. Studies on a hemolytic factor of cobra venom requiring a heat-labile serum factor. Biochim Biophys Acta. 1970;201:364–74. doi: 10.1016/0304-4165(70)90311-9. [DOI] [PubMed] [Google Scholar]
- 194.Müller-Eberhard H, Fjellström K. Isolation of the anticomplementary protein from cobra venom and its mode of action on C3. J Immunol. 1971;107:1666–72. [PubMed] [Google Scholar]
- 195.Cooper N. Formation and function of a complex of the C3 proactivator with a protein from cobra venom. J Exp Med. 1973;137:451–60. doi: 10.1084/jem.137.2.451. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Weiler J, Yurt R, Fearon D, et al. Modulation of the formation of the amplification convertase of complement, C3b, Bb, by native and commercial heparin. J Exp Med. 1978;147:409–21. doi: 10.1084/jem.147.2.409. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Baker P, Lint T, McLeod B, et al. Studies on the inhibition of C56-induced lysis (reactive lysis). VI. Modulation of C56-induced lysis polyanions and polycations. J Immunol. 1975;114:554–8. [PubMed] [Google Scholar]
- 198.Hughes-Jones N, Gardner B. The reaction between the complement subcomponent C1q, IgG complexes and polyionic molecules. Immunology. 1978;34:459–63. [PMC free article] [PubMed] [Google Scholar]
- 199.Almeda S, Rosenberg R, Bing D. The binding properties of human complement component C1q. Interaction with mucopolysaccharides. J Biol Chem. 1983;258:785–91. [PubMed] [Google Scholar]
- 200.Weiler J. Polyions regulate the alternative amplification pathway of complement. Immunopharmacology. 1983;6:245–55. doi: 10.1016/0162-3109(83)90024-3. [DOI] [PubMed] [Google Scholar]
- 201.Weiler J, Edens R, Linhardt R, et al. Heparin and modified heparin inhibit complement activation in vivo. J Immunol. 1992;148:3210–5. [PubMed] [Google Scholar]
- 202.Edens R, Linhardt R, Bell C, et al. Heparin and derivatized heparin inhibit zymosan and cobra venom factor activation of complement in serum. Immunopharmacology. 1994;27:145–53. doi: 10.1016/0162-3109(94)90049-3. [DOI] [PubMed] [Google Scholar]
- 203.Bergamaschini L, Rossi E, Storini C, et al. Peripheral treatment with enoxaparin, a low molecular weight heparin, reduces plaques and beta-amyloid accumulation in a mouse model of Alzheimer’s disease. J Neurosci. 2004;24:4181–6. doi: 10.1523/JNEUROSCI.0550-04.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204.Asghar S. Pharmacological manipulation of complement system. Pharmacol Rev. 1984;36:223–44. [PubMed] [Google Scholar]
- 205.Hong K, Kinoshita T, Kitajima H, et al. Inhibitory effect of K-76 monocarboxylic acid, an anticomplementary agent, on the C3b inactivator system. J Immunol. 1981;127:104–8. [PubMed] [Google Scholar]
- 206.Miyazaki W, Izawa T, Nakano Y, et al. Effects of K-76 monocarboxylic acid, an anticomplementary agent, on various in vivo immunological reactions and on experimental glomerulonephritis. Complement. 1984;1:134–46. doi: 10.1159/000467829. [DOI] [PubMed] [Google Scholar]
- 207.Englberger W, Hadding U, Etschenberg E, et al. Rosmarinic acid: a new inhibitor of complement C3-convertase with anti-inflammatory activity. Int J Immunopharmacol. 1988;10:729–37. doi: 10.1016/0192-0561(88)90026-4. [DOI] [PubMed] [Google Scholar]
- 208.Sahu A, Rawal N, Pangburn M. Inhibition of complement by covalent attachment of rosmarinic acid to activated C3b. Biochem Pharmacol. 1999;57:1439–46. doi: 10.1016/s0006-2952(99)00044-1. [DOI] [PubMed] [Google Scholar]
- 209.Ling M, Piddlesden S, Morgan B. A component of the medicinal herb ephedra blocks activation in the classical and alternative pathways of complement. Clin Exp Immunol. 1995;102:582–8. doi: 10.1111/j.1365-2249.1995.tb03856.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 210.Fujii S, Hitomi Y. New synthetic inhibitors of C1r, C1 esterase, thrombin, plasmin, kallikrein and trypsin. Biochim Biophys Acta. 1981;661:342–5. doi: 10.1016/0005-2744(81)90023-1. [DOI] [PubMed] [Google Scholar]
- 211.Inagi R, Miyata T, Maeda K, et al. FUT-175 as a potent inhibitor of C5/C3 convertase activity for production of C5a and C3a. Immunol Lett. 1991;27:49–52. doi: 10.1016/0165-2478(91)90243-4. [DOI] [PubMed] [Google Scholar]
- 212.Ino Y, Sato T, Koshiyama Y, et al. Effects of FUT-175, a novel synthetic protease inhibitor, on the development of adjuvant arthritis in rats and some biological reactions dependent on complement activation. Gen Pharmacol. 1987;18:513–6. doi: 10.1016/0306-3623(87)90072-3. [DOI] [PubMed] [Google Scholar]
- 213.Miyagawa S, Shirakura R, Matsumiya G, et al. Prolonging discordant xenograft survival with anticomplement reagents K76COOH and FUT175. Transplantation. 1993;55:709–13. doi: 10.1097/00007890-199304000-00004. [DOI] [PubMed] [Google Scholar]
- 214.Holland M, Morikis D, Lambris J. Synthetic small-molecule complement inhibitors. Curr Opin Investig Drugs. 2004;5:1164–73. [PubMed] [Google Scholar]
- 215.Morgan B, Harris C. Complement therapeutics; history and current progress. Mol Immunol. 2003;40:159–70. doi: 10.1016/s0161-5890(03)00111-1. [DOI] [PubMed] [Google Scholar]
- 216.Weisman H, Bartow T, Leppo M, et al. Soluble human complement receptor type 1: in vivo inhibitor of complement suppressing post-ischemic myocardial inflammation and necrosis. Science. 1990;249:146–51. doi: 10.1126/science.2371562. [DOI] [PubMed] [Google Scholar]
- 217.Lazar H, Keilani T, Fitzgerald C, et al. Beneficial effects of complement inhibition with soluble complement receptor 1 (TP10) during cardiac surgery: is there a gender difference. Circulation. 2007;116:I83–8. doi: 10.1161/CIRCULATIONAHA.106.677914. [DOI] [PubMed] [Google Scholar]
- 218.Lazar H, Bokesch P, Van Lenta F, et al. Soluble human complement receptor 1 limits ischemic damage in cardiac surgery patients at high risk requiring cardiopulmonary bypass. Circulation. 2004;110:II274–9. doi: 10.1161/01.CIR.0000138315.99788.eb. [DOI] [PubMed] [Google Scholar]
- 219.Harris C, Williams A, Linton S, et al. Coupling complement regulators to immunoglobulin domains generates effective anti-complement reagents with extended half-life in vivo. Clin Exp Immunol. 2002;129:198–207. doi: 10.1046/j.1365-2249.2002.01924.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220.Smith G, Smith R. Membrane-targeted complement inhibitors. Mol Immunol. 2001;38:249–55. doi: 10.1016/s0161-5890(01)00047-5. [DOI] [PubMed] [Google Scholar]
- 221.Song H, He C, Knaak C, et al. Complement receptor 2-mediated targeting of complement inhibitors to sites of complement activation. J Clin Invest. 2003;111:1875–85. doi: 10.1172/JCI17348. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222.Carrier M, Ménasché P, Levy J, et al. Inhibition of complement activation by pexelizumab reduces death in patients undergoing combined aortic valve replacement and coronary artery bypass surgery. J Thorac Cardiovasc Surg. 2006;131:352–6. doi: 10.1016/j.jtcvs.2005.10.011. [DOI] [PubMed] [Google Scholar]
- 223.Brodsky R, Young N, Antonioli E, et al. Multicenter phase 3 study of the complement inhibitor eculizumab for the treatment of patients with paroxysmal nocturnal hemoglobinuria. Blood. 2008;111:1840–7. doi: 10.1182/blood-2007-06-094136. [DOI] [PubMed] [Google Scholar]
- 224.Kaplan M. Eculizumab (Alexion) Curr Opin Investig Drugs. 2002;3:1017–23. [PubMed] [Google Scholar]
- 225.Bora N, Kaliappan S, Jha P, et al. CD59, a complement regulatory protein, controls choroidal neovascularization in a mouse model of wet-type age-related macular degeneration. J Immunol. 2007;178:1783–90. doi: 10.4049/jimmunol.178.3.1783. [DOI] [PubMed] [Google Scholar]
- 226.Zhang H, Yu J, Bajwa E, et al. Targeting of functional antibody-CD59 fusion proteins to a cell surface. J Clin Invest. 1999;103:55–61. doi: 10.1172/JCI4607. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227.Schenk D, Barbour R, Dunn W, et al. Immunization with amyloid-beta attenuates Alzheimer-disease-like pathology in the PDAPP mouse. Nature. 1999;400:173–7. doi: 10.1038/22124. [DOI] [PubMed] [Google Scholar]
- 228.Janus C, Pearson J, McLaurin J, et al. A beta peptide immunization reduces behavioural impairment and plaques in a model of Alzheimer’s disease. Nature. 2000;408:979–82. doi: 10.1038/35050110. [DOI] [PubMed] [Google Scholar]
- 229.DeMattos R, Bales K, Cummins D, et al. Peripheral anti-A beta antibody alters CNS and plasma A beta clearance and decreases brain A beta burden in a mouse model of Alzheimer’s disease. Proc Natl Acad Sci USA. 2001;98:8850–5. doi: 10.1073/pnas.151261398. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 230.Dodart J, Bales K, Gannon K, et al. Immunization reverses memory deficits without reducing brain Abeta burden in Alzheimer’s disease model. Nat Neurosci. 2002;5:452–7. doi: 10.1038/nn842. [DOI] [PubMed] [Google Scholar]
- 231.Wisniewski T, Frangione B. Immunological and anti-chaperone therapeutic approaches for Alzheimer disease. Brain Pathol. 2005;15:72–7. doi: 10.1111/j.1750-3639.2005.tb00102.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 232.Bayer A, Bullock R, Jones R, et al. Evaluation of the safety and immunogenicity of synthetic Abeta42 (AN1792) in patients with AD. Neurology. 2005;64:94–101. doi: 10.1212/01.WNL.0000148604.77591.67. [DOI] [PubMed] [Google Scholar]
- 233.Pride M, Seubert P, Grundman M, et al. Progress in the active immunotherapeutic approach to Alzheimer’s disease: clinical investigations into AN1792-associated meningoencephalitis. Neurodegener Dis. 2008;5:194–6. doi: 10.1159/000113700. [DOI] [PubMed] [Google Scholar]
- 234.Orgogozo J, Gilman S, Dartigues J, et al. Subacute meningoencephalitis in a subset of patients with AD after Abeta42 immunization. Neurology. 2003;61:46–54. doi: 10.1212/01.wnl.0000073623.84147.a8. [DOI] [PubMed] [Google Scholar]
- 235.Boche D, Nicoll J. The role of the immune system in clearance of Abeta from the brain. Brain Pathol. 2008;18:267–78. doi: 10.1111/j.1750-3639.2008.00134.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 236.Boche D, Zotova E, Weller R, et al. Consequence of Abeta immunization on the vasculature of human Alzheimer’s disease brain. Brain. 2008;131:3299–310. doi: 10.1093/brain/awn261. [DOI] [PubMed] [Google Scholar]
- 237.Bombois S, Maurage C, Gompel M, et al. Absence of beta-amyloid deposits after immunization in Alzheimer disease with Lewy body dementia. Arch Neurol. 2007;64:583–7. doi: 10.1001/archneur.64.4.583. [DOI] [PubMed] [Google Scholar]
- 238.Masliah E, Hansen L, Adame A, et al. Abeta vaccination effects on plaque pathology in the absence of encephalitis in Alzheimer disease. Neurology. 2005;64:129–31. doi: 10.1212/01.WNL.0000148590.39911.DF. [DOI] [PubMed] [Google Scholar]
- 239.Nicoll J, Barton E, Boche D, et al. Abeta species removal after abeta42 immunization. J Neuropathol Exp Neurol. 2006;65:1040–8. doi: 10.1097/01.jnen.0000240466.10758.ce. [DOI] [PubMed] [Google Scholar]
- 240.Hock C, Konietzko U, Papassotiropoulos A, et al. Generation of antibodies specific for beta-amyloid by vaccination of patients with Alzheimer disease. Nat Med. 2002;8:1270–5. doi: 10.1038/nm783. [DOI] [PubMed] [Google Scholar]
- 241.Nicoll J, Wilkinson D, Holmes C, et al. Neuropathology of human Alzheimer disease after immunization with amyloid-beta peptide: a case report. Nat Med. 2003;9:448–52. doi: 10.1038/nm840. [DOI] [PubMed] [Google Scholar]
- 242.Webster S, Galvan M, Ferran E, et al. Antibody-mediated phagocytosis of the amyloid beta-peptide in microglia is differentially modulated by C1q. J Immunol. 2001;166:7496–503. doi: 10.4049/jimmunol.166.12.7496. [DOI] [PubMed] [Google Scholar]
- 243.Sadowski M, Wisniewski T. Disease modifying approaches for Alzheimer’s pathology. Curr Pharm Des. 2007;13:1943–54. doi: 10.2174/138161207781039788. [DOI] [PubMed] [Google Scholar]
- 244.Gilman S, Koller M, Black R, et al. Clinical effects of Abeta immunization (AN1792) in patients with AD in an interrupted trial. Neurology. 2005;64:1553–62. doi: 10.1212/01.WNL.0000159740.16984.3C. [DOI] [PubMed] [Google Scholar]
- 245.Holmes C, Boche D, Wilkinson D, et al. Long-term effects of Abeta42 immunisation in Alzheimer’s disease: follow-up of a randomised, placebo-controlled phase I trial. Lancet. 2008;372:216–23. doi: 10.1016/S0140-6736(08)61075-2. [DOI] [PubMed] [Google Scholar]
- 246.Wisniewski T. AD vaccines: conclusions and future directions. CNS Neurol Disord Drug Targets. 2009;8:160–6. doi: 10.2174/187152709787847289. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 247.Rakover I, Arbel M, Solomon B. Immunotherapy against APP beta-secretase cleavage site improves cognitive function and reduces neuroinflammation in Tg2576 mice without a significant effect on brain abeta levels. Neurodegener Dis. 2007;4:392–402. doi: 10.1159/000103250. [DOI] [PubMed] [Google Scholar]
- 248.Wilk S, Chen W. Synthetic peptide-based activators of the proteasome. Mol Biol Rep. 1997;24:119–24. doi: 10.1023/a:1006851428691. [DOI] [PubMed] [Google Scholar]
- 249.Wilk S, Chen W, Magnusson R. Modulation of the PA28 alpha-20S proteasome interaction by a peptidyl alcohol. Arch Biochem Biophys. 1999;362:283–90. doi: 10.1006/abbi.1998.1034. [DOI] [PubMed] [Google Scholar]
- 250.Demena I, Mahillo E, Arribas J, et al. Kinetic mechanism of activation by cardiolipin (diphosphatidylglycerol) of the rat liver multicatalytic proteinase. Biochem J. 1993;296:93–7. doi: 10.1042/bj2960093. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 251.Xie Y, Varshavsky A. RPN4 is a ligand, substrate, and transcriptional regulator of the 26S proteasome: A negative feedback circuit. Proc Natl Acad Sci USA. 2001;98:3056–61. doi: 10.1073/pnas.071022298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 252.Rockenstein E, Torrance M, Mante M, et al. Cerebrolysin decreases amyloid-beta production by regulating amyloid protein precursor maturation in a transgenic model of Alzheimer’s disease. J Neurosci Res. 2006;83:1252–61. doi: 10.1002/jnr.20818. [DOI] [PubMed] [Google Scholar]
- 253.Rockenstein E, Mallory M, Mante M, et al. Effects of Cerebrolysin on amyloid-beta deposition in a transgenic model of Alzheimer’s disease. J Neural Transm Suppl. 2002;62:327–36. [PubMed] [Google Scholar]
- 254.Rüther E, Ritter R, Apecechea M, et al. Sustained improvements in patients with dementia of Alzheimer’s type (DAT) 6 months after termination of Cerebrolysin therapy. J Neural Transm. 2000;107:815–29. doi: 10.1007/s007020070061. [DOI] [PubMed] [Google Scholar]
- 255.Melnikova I. Therapies for Alzheimer’s disease. Nat Rev Drug Discov. 2007;6:341–2. doi: 10.1038/nrd2314. [DOI] [PubMed] [Google Scholar]
- 256.Lleó A, Greenberg S, Growdon J. Current pharmacotherapy for Alzheimer’s disease. Annu Rev Med. 2006;57:513–33. doi: 10.1146/annurev.med.57.121304.131442. [DOI] [PubMed] [Google Scholar]
- 257.Bureeva S, Andia-Pravdivy J, Kaplun A. Drug design using the example of the complement system inhibitors’ development. Drug Discov Today. 2005;10:1535–42. doi: 10.1016/S1359-6446(05)03592-0. [DOI] [PubMed] [Google Scholar]
- 258.Makrides S. Therapeutic inhibition of the complement system. Pharmacol Rev. 1998;50:59–87. [PubMed] [Google Scholar]
