Abstract
As the most prevalent form of dementia worldwide, Alzheimer's disease (AD) continues to be a burden for patients and their families. In addition, with the global population of aged individuals increasing exponentially, AD represents a signifcant economic burden to society. The development of an effective approach for the treatment of AD is thus of major importance, as current treatment strategies are limited to agents that attenuate disease symptomatology without addressing the causes of disease. A considerable need exists for the development of an effective therapy to prevent, or at least delay, the progression of AD. Current hypotheses for the pathogenesis of AD are discussed in this review, with a particular emphasis on the implications of these hypotheses with respect to treatment strategies and preventive measures.
Keywords: Aβ modulator, Alzheimer's disease, antioxidant, cholinesterase inhibitor, excitotoxicity, mitochondrial dynamics, neurofibrillary tangle, oxidative stress, tau, therapeutic
Introduction
The complex molecular interactions that are involved in the development and progression of Alzheimer's disease (AD) are only partly understood. While early-onset familial AD is linked directly to specific gene mutations, the late-onset sporadic form of the disease, which accounts for approximately 95% of all AD cases [1], likely results from the complex interplay of molecular, environmental and genetic factors. With the identification of novel mechanisms that lead to neurodegeneration, the characteristic pathologies of AD, notably the β-amyloid (Aβ)-containing senile plaques and the hyperphosphorylated tau protein aggregates that form neurofibrillary tangles (NFTs), are no longer considered to be causative but, instead, are considered to be the secondary products of prior insults [2]. Importantly, the development of NFTs may protect against neuronal damage, as mutations in the Aβ precursor protein (APP) [3–5] and the presence of NFT-containing neurons [6,7] are associated with the reduced steady-state production of Aβ [8] and with reduced levels of oxidative stress, respectively [9]. Moreover, rather than readily succumbing to neuronal death, NFT-containing neurons often survive for decades [10]. Conversely, oxidative stress and mitochondrial malfunction are two of the primary factors that are believed to be responsible for initiating the neuronal cascade that leads to AD [11,12]. Without intervention, oxidative stress and mitochondrial malfunction may elicit the production and aggregation of Aβ and tau, as well as neuroinflammation, aberrations in the cell cycle, small-vessel disease and an overall decline in cognitive function [12,13].
Notably, the aggregation of Aβ in the form of senile plaques and the formation and fibrillization of hyperphosphorylated tau into NFTs generate strong responses from the surrounding cellular environment and are responsible for much of the late-stage cognitive decline observed in patients with AD. Alternative theories for the pathogenesis of AD suggest that such pathologies are primarily responsible for neurodegeneration [14]. For example, once Aβ has aggregated sufficiently, this protein elicits (among other effects) a neuroinflammatory response via the activation of microglia and astrocytes [15,16]. Following the initial neuroinflammatory response, the neurotoxic biproducts of inflammation, such as the downstream intermediates of the kynurenine pathway (the primary route of l-tryptophan metabolism), cause additional oxidative damage to cells [17–23]. Similarly, the hyperphosphorylated tau fibrils create cytoskeletal stresses and promote neuronal dysfunction [24,25]. Therefore, the chronology and causality of the multitude of pathologies observed in AD are responsible for instilling controversy in the field, rather than the effects of such pathologies.
Three approaches for the treatment of AD are possible: (i) treatments that prevent the onset of the disease by sequestering the primary progenitors; (ii) disease-modifying treatments that target and reduce the secondary pathologies of the disease, leading to the cessation or the reversal of disease progression; and (iii) symptomatic treatments that treat the tertiary cognitive symptoms of the disease and protect from further cognitive decline. Notably, the appropriate treatment method depends on the severity of the disease in each individual; however, preventive and disease-modifying treatment strategies are the only hope for the eradication of AD from the general population.
Preventive treatments for Alzheimer's disease
The prevention of AD is the most effective strategy for eliminating the occurrence of this disease. Oxidative stress and mitochondrial abnormalities have been implicated in the initiation of AD [9,26,27]; a successful prevention regimen thus may act by attenuating these phenomena [28].
Antioxidants
Oxidative stress represents a particularly attractive target for therapeutic intervention because of the ubiquitous role that this type of stress plays in disease development. Reactive oxidative species (ROS) are generated within the mitochondria of normal respiring cells [29]: the oxidation-reduction reactions that establish the proton gradient in oxidative phosphorylation (OXPHOS)/ATP production generate free radical intermediates via the transfer of electrons. Despite the resident cellular sequestration mechanisms that antagonize the release of ROS, free radicals escape from the electron transport chain (ETC; the escape of free radicals is estimated to be approximately 1011 ROS/cell/day [29]), causing damage to mitochondrial DNA (mtDNA), proteins and phospholipids. The levels of damaged mtDNA, proteins and phospholipids are increased in AD [2,11,13], suggesting that antioxidant therapies may be protective against free-radical generation and cellular oxidative damage in this disease.
Naturally occurring antioxidants represent one treatment option for AD. These compounds, including vitamin E and vitamin C, are associated with the reduced prevalence and incidence of AD [30–32] and, when used in combination, are considered to have synergistic biochemical antioxidant potential. However, vitamin E treatment alone was not effective against the progression of AD in randomized clinical trials [33].
Mitochondria are the primary source of ROS [34], and oxidative damage to mitochondrial components precedes that of any other cellular component during the development of neurodegenerative disease [13,35–37]. Two potential antioxidant drugs that are targeted to the mitochondria are under investigation for the potential treatment of AD. The first drug is coenzyme Q10 (CoQ10), a component of the ETC that is present in the inner mitochondrial membrane and conducts the high-energy electrons from complex I to complex II. Several studies have demonstrated that nullifying oxidative damage and attenuating mitochondrial dysfunction has neuroprotective effects [38,39]. For example, in an MC65 neuroblastoma cell model of AD, CoQ10 suppressed H2O2 and O2- production (the most prevalent free radicals) and provided complete protection from oxidative-induced neurotoxicity [40]. However, CoQ10 has two major limitations as a therapy. First, the functioning of CoQ10 is dependent entirely on the functioning of the ETC, which facilitates the redox cycling of the enzyme from ubiquinone (oxidized form) to ubiquinol (reduced form). This requirement becomes critical when oxidatively damaged mitochondria contain defective OXPHOS systems and damaged ETCs, and thus cannot recycle CoQ10 appropriately. Second, although oxidative damage was attenuated following the oral administration of CoQ10 to rodents, the levels of the CoQ10 protein were not increased in the brain [40–42], suggesting that CoQ10 is unable to cross the blood-brain barrier (BBB). Thus, CoQ10 derivatives are being developed that do not require intact ETC functions and that demonstrate increased BBB permeability.
The second potential antioxidant drug is the CoQ10 derivative MitoQ (Antipodean Pharmaceuticals Inc), a triphenylphosphonium-linked ubiquinone derivative [43]. MitoQ concentrates highly in the mitochondria because of the large mitochondrial membrane potential (Δψm) [44], and is also an effective antioxidant in the absence of a functioning ETC [45,46]. For example, in in vitro studies using a leukemic cell model, following the depletion of glutathione, MitoQ blocked the generation of ROS effectively, protected the mitochondrial protein redox status, preserved the structural integrity of mitochondria, and blocked cell death [45]. MitoQ is in phase II clinical trials for Parkinson's disease and liver damage associated with HCV infection [47].
Other mitochondrial antioxidants that are under investigation for the potential treatment of AD include acetyl-l-carnitine (ALCAR) and r-α-lipoic acid (LA), both of which were demonstrated to reduce oxidative stress and mitochondrial abnormalities in cellular mouse models of AD [48], and to restore cognitive functions in aged rats [49–53] and dogs [54,55]. Notably, the coadministration of ALCAR and LA reduced the level of damage to hippocampal neuronal mitochondria significantly, with the mitochondria retaining their structural integrity and the number of normal mitochondria increasing significantly in both young and old rats [52]. These results provide considerable hope for the effectiveness of future therapies that target oxidative stress and mitochondrial disruptions in AD.
Protective agents of mitochondrial dynamics
Mitochondrial integrity is vital to cellular health. Mitochondria are not static organelles, but are dynamic bodies that divide constantly and fuse within the cell in response to environmental demands [56,57]. The delicate balance of fission and fusion is highly regulated by specific mitochondrial membrane proteins and their associated complexes [12,56], and provides, among other functions, a defensive mechanism against the deleterious presence of defective mitochondria that may be the result of mutation or oxidative damage. Mitochondrial fusion permits the exchange of lipid membranes and inter-mitochondrial contents (ie, mtDNA, and OXPHOS and ETC proteins); and mitochondrial fission, coupled with mitochondrial fusion and autophagy, allows the sequestration and elimination of irreversibly damaged mitochondria [58–60]. The neurons of patients with AD demonstrate significant alterations in mitochondrial structure [61,62]. In addition, the mitochondrial distribution and levels of fission and fusion proteins are altered in hippocampal pyramidal neurons in patients with AD [63,64]. Therefore, an agent that protects the proteins that are involved in mitochondrial fission and fusion may maintain the protective balance of these proteins and may prevent the neurodegeneration observed in AD.
Notably, while preventive measures would protect the brain from the onset of AD, after significant oxidative damage and mitochondrial imbalances have occurred and the associated pathologies have accumulated within the brain, reversing the course of the disease is likely beyond the scope of agents that target oxidative damage and modulate mitochondrial dynamics. At that point in disease progression, only disease-modifying or symptomatic treatments would be of benefit to patients.
Disease-modifying treatments for Alzheimer's disease
The neuropathological cascade of AD results from the age-related accumulation of neurodegenerative initiators and mediators, such as ROS and mtDNA mutations. However, after a threshold of cellular damage has been reached, the cell enters a compensatory 'oxidative steady state' that enables a basic level of operational activity, despite the depleted levels of functioning mitochondria and other cellular components. The hallmark pathologies of AD develop during this period of steady-state oxidation, and medications targeting such secondary pathologies may inhibit further disease progression.
Anti-Aβ therapies
Interestingly, research indicates that Aβ secretion has antioxidant properties [65–71] and is a compensatory strategy that is used by the cell to antagonize uncontrolled oxidative stress [2,11,13]. In AD, Aβ secretion follows the appearance of ROS and mitochondrial anomalies [29]. However, the continuous secretion of Aβ, coupled with the continuous levels of oxidative stress that are caused by mitochondrial damage, contributes to the cascade of events that lead to neurodegeneration. In particular, Aβ can succumb to oxidative stress and develop dityrosine cross-linkages that prevent the protein from being soluble in vivo, thereby promoting Aβ aggregation [70,72]. This non-soluble, antagonistic form of Aβ and the formation of senile plaques are the primary targets for the therapeutic strategies that are being investigated for AD.
Three treatment approaches that target Aβ may provide potential benefits to patients with AD: anti-aggregation agents; secretase enzyme modulators; and Aβ-clearance mechanisms. These approaches are described in the following sections.
Anti-Aβ aggregation
The anti-aggregation agents target the fibrillization of the Aβ peptide. One anti-aggregation approach that has demonstrated significant results in vitro is that of nanoparticle-conjugated metal chelators. Chelators disrupt the interactions of Aβ with redox metals in the brain, thereby preventing aggregation. Specifically, iron and copper, the levels of which are increased in the brains of patients with AD [73,74], as well as zinc, are suggested to play an important role in Aβ self-assembly and neurotoxicity [75–78]. While metal chelators (eg, desferrioxamine, EDTA [ethylenediaminetetracetic acid] and clioquinol [iodochlorohydroxyquin] [79–81]) can attenuate the effects of Aβ [71,82–84], their use as therapeutic agents is hindered by an inability to cross the BBB. However, a recent study proposed the use of nanoparticle conjugation to deliver iron chelators effectively across the BBB, without affecting their metal-binding ability [85,86]. In this study, the Nano-N2PY nanoparticle-chelator conjugate protected neuronal cells against Aβ-associated cyctotoxicity in vitro, with no adverse effects on cell growth or proliferation. Furthermore, the conjugate prevented the formation of Aβ-aggregate completely [85]. These results have yet to be translated to animal models of neurodegeneration, but present exciting potential for the use of metal chelators as therapies for AD.
The aggregation of Aβ was also diminished significantly in transgenic (Tg) mouse models of AD following the administration of the neurohormone melatonin [87]. The long-term administration of melatonin reduced the deposition of Aβ within the hippocampus and entorhinal cortex significantly [87], presumably through the prevention of Aβ fibrillogenesis [88–90]. Although melatonin also suppressed the levels of proinflammatory cytokines (eg, TNFα) and reduced the levels of oxidative stress in the cortex, its primary mode of action was the prevention of Aβ aggregation. As aggregated Aβ itself induces inflammatory neuronal responses, as well as ROS generation and mitochondrial damage, the effects of melatonin on oxidative stress and proinflammatory cytokine levels may be the result of its suppression of Aβ aggregation. Moreover, the long-term safety of melatonin in humans is known, and this fact, when combined with the beneficial attenuation of the pathogenesis of AD, renders this agent particularly appealing.
The cyclohexanehexol stereoisomer ELND-005 (scyllo-inositol; Elan Corp plc/Transition Therapeutics Inc) is also under investigation for its anti-Aβ oligomerization properties [91]. The administration of ELND-005 to Tg mice was successful in reducing the accumulation of Aβ oligomers, and also reversed memory deficits [92]. Phase II clinical trials of the compound are ongoing, with results expected by mid-2010 (ClinicalTrials.gov identifiers: NCT00568776 and NCT00934050).
Secretase enzyme modulation
Another approach for reducing the levels of Aβ involves modulating the activity of the β- and γ-secretase cleaving enzymes to inhibit the production of Aβ. The β- and γ-secretase enzymes are aspartyl proteases that convert APP to Aβ; treatment strategies that involve the inhibition of these two enzymes aim to reduce the levels of cerebral amyloid [93,94]. Currently, several agents targeting these enzymes are being developed, including the γ-secretase inhibitors MK-0752 (Merck & Co Inc) and semagacestat (LY-450139; Eli Lilly & Co; NCT00762411 and NCT00594568) [94–96], and the β-secretase inhibitor Posiphen (QR Pharma Inc) [91,97]. Phase II clinical trials for Posiphen are ongoing [91,98], and semagacestat has progressed to phase III trials (NCT01035138).
Aβ clearance
Strategies that aim to reduce the cerebral levels of Aβ directly are of therapeutic significance; immunotherapy is one such approach that is at advanced stages of clinical testing for AD, despite the potential safety concerns of this approach [99,100]. Immunotherapies are designed to use the body's immune system to eliminate the deposition of existing Aβ senile plaques. Active immunization requires the introduction of an antigen that mimics Aβ, so that antibodies targeting the antigen are produced; these antibodies subsequently bind to Aβ to facilitate its clearance from the CNS [93,97]. The development of AN-1792 (an aggregated amyloid peptide), the first active immunotherapy for AD, was halted in January 2002 because of the occurrence of meningoencephalitis in 6% of vaccinated patients. Second–generation amyloid antigens have been designed to decrease the likelihood of T-cell activation, which was considered responsible for the adverse affects observed with AN-1792 [93]. These second-generation immunotherapies include CAD-106 (Cytos Biotechnology AG/Novartis AG) and vanutide cridificar (ACC-001; JANSSEN Alzheimer Immunotherapy/Pfizer Inc), both of which are in phase II clinical trials (eg, NCT00956410 and NCT00479557, respectively).
Alternatively, the passive immunization method involves the parenteral administration of a specific antibody or pooled non-specific antibodies, thus requiring less contribution from the patient's immune response – a major limitation in an elderly cohort. One such passive immunization approach is the humanized Aβ-targeted mAb bapineuzumab (AAB-001; JANSSEN Alzheimer Immunotherapy/Pfizer Inc), which is in phase III clinical trials in patients with AD [91]; however, treatment with this mAb has not resulted in improvements in cognitive performance tests (Alzheimer's Disease Assessment Scale Cognitive Subscale [ADAS-cog]), except in post-hoc analyses (NCT00667810, NCT00676143, NCT00575055 and NCT00574132) [101]. The phase III trial, which is expected to be completed in April 2011, will elaborate on the safety and efficacy of bapineuzumab [91]. Other passive immunization drugs that are being developed include solanezumad (LY-2062430; Eli Lilly), which has completed phase II trials (NCT00329082, NCT00749216 and NCT00749216) and is undergoing phase III trials (NCT00905372 and NCT00904683); an immunoglobulin (Baxter International) that is in phase II trials [102]; and PF-4360365 (PF-04360365; Pfizer), which is undergoing phase II trials (NCT00722046).
Cholesterol-lowering statins, such as the FDA-approved drug simvastatin [103], have been clinically unsuccessful as therapies for AD, despite demonstrating initial Aβ clearance effects [93,97]. Therefore, whether amyloid-lowering strategies ultimately will be successful in the treatment of AD is unclear [99,100].
Tau-based therapies
Another significant aspect of AD pathology that provides a potential target for therapeutic intervention is the hyperphosphorylated form of the microtubule-associated protein tau. Tau hyperphosphorylation and the presence of this protein in an aggregated form in NFTs are correlated with cognitive decline in patients with AD [8,104]. Furthermore, NFT concentrations are used as a definitive marker for the postmortem diagnosis of AD [105]. Thus, therapeutic strategies that target hyperphosphorylated tau proteins are potentially relevant for the treatment of AD.
Microtubule stabilization
During the past several years, the investigation of NFTs as a possible therapeutic target for the treatment of AD has yielded several exciting approaches. First, as the hyperphosphorylation and subsequent aggregation of tau in NFTs impairs the function of microtubules within the cell [24,25], and reduces microtubule density and fast axonal transport [106,107], an agent that stabilizes microtubules in vivo has potential therapeutic efficacy in AD. In particular, the administration of the microtubule-stabilizing drug paclitaxel to tau-Tg mice for a 3-month period resulted in significant improvements in fast axonal transport and microtubule density [108]. Motor function was also improved in paclitaxel-treated mice [108]. Importantly, however, paclitaxel does not cross the BBB readily [109,110], and must therefore reach the brain indirectly via a peripheral uptake and retrograde transport mechanism, a requirement that reduces the efficacy of this drug considerably [111]. The limited ability of paclitaxel to cross the BBB presents a significant challenge because the high doses required for this drug to enter the brain elicit a potent antimitotic element [111] and other adverse side effects [112]. Fortunately, the octapeptide NAPVSIPQ, termed NAP based on the amino acid sequence, can cross the BBB and was demonstrated to reduce tau phosphorylation in tau-Tg mice, presumably via the stabilization of microtubules [109,113,114]; the effect of NAP provides support for the possibility of an effective microtubule-stabilizing drug for the treatment of AD.
Inhibition of hyperphosphorylation
The attenuation of NFT formation may also be achieved via the inhibition of tau hyperphosphorylation. Specifically, the phosphorylation of the tau protein at Ser262, Ser235 and Thr231 was demonstrated to enhance tau fibrillization and aggregation at the expense of the microtubules with which the protein associates [115–118]. Therefore, methods aimed at preventing the phosphorylation of specific sites in the tau protein may elicit beneficial results, particularly if the effect of the inhibitor were sufficiently selective. In this regard, glycogen synthase kinase 3 (GSK3), an enzyme that is responsible for the regulation of various cell processes [119], was demonstrated to colocalize with NFTs in the brains of patients with AD [120]. In addition, the overexpression of GSK3 in Tg mice resulted in increased tau hyperphosphorylation and behavioral deficits [121,122]. Significantly, the administration of lithium chloride (an inhibitor of GSK3) to tau-Tg mice reduced the levels of both tau phosphorylation and insoluble tau [123,124]. In addition, the treatment of presymptomatic animals with lithium chloride prevented the onset of tau pathology and reduced existing pathologies in mice with NFTs, although the presence of NFTs persisted following treatment [125]. Similarly, in rodent models, other small-molecule tau-kinase inhibitors were demonstrated to reduce tau hyperphosphorylation, including the non-specific kinase inhibitor SRN-003–556 and the GSK3 inhibitors CHIR-98014 and SB-216763 [126].
The tau protein may also be modified post-translationally at certain serine and threonine residues via the addition of β-N-acetylglucosamine (O-GlcNAc), in a manner that is reciprocal to tau phosphorylation and thus to fibrillar formation. Consequently, increased levels of tau O-linked glycosylation correspond directly to decreased levels of tau phosphorylation [127,128]. Therefore, the modulation of the O-GlcNAc-cleaving enzyme O-GlcNAcase has the potential to prevent tau hyperphosphorylation. Consistent with this hypothesis, the acute administration of the O-GlcNAcase inhibitor thiamet-G to normal rats reduced the phosphorylation of tau at the Ser396, Ser404 and Thr231 residues [127], thereby demonstrating the mechanistic importance of O-GlcNAcase in tau phosphorylation.
Inhibition of oligomerization/fibrillization
Another method of disrupting NFTs involves the direct inhibition of tau oligomers and of fibril assembly that prevents the formation of toxic species and increases the levels of monomeric tau, leading to the effective stabilization of cellular microtubules [111,129]. The histological dye methylene blue (Rember; TauRx Therapeutics Pte Ltd) was reported to inhibit tau interactions and to alter the structure of the paired helical filaments of tau in brains from patients with AD. Phase II clinical trials of methylene blue have been completed with therapeutically positive data, and phase III trials are planned [97,130]. Similar inhibitors of tau-fibril assembly have been identified and are being investigated, but none has reached the clinical stages of development [131–133].
Enhancement of tau degradation
The disruptive effects of aggregated, hyperphosphorylated tau can also be eliminated by the upregulation of the intracellular degradation of the protein through the ubiquitin proteosome system or through macroautophagy [111]. In the ubiquitin proteosome degradation pathway, a targeted protein is tagged with ubiquitin and subsequently recognized and degraded by the proteosome complex [134]. As the ubiquitin proteosome system requires that the target protein is threaded through the narrow opening of the proteosome, the activation of this system degrades only the non-fibrillar phosphorylated tau. Nevertheless, the Hsp90 inhibitor mediated degradation of the smaller non-fibrillar phosphorylated tau. Because Hsp90 is primarily responsible for the ATP-driven refolding of denatured proteins, the inhibition of this protein halts the attempted preservation of phosphorylated tau by this chaperone effectively, thereby enhancing tau degradation [135–137]. For example, the Hsp90 inhibitor EC-102, which was administered to human tau-expressing Tg mice for 7 days, reduced the levels of hyperphosphorylated tau in the brain [135,138]. Moreover, EC-102 inhibited the formation of Hsp90/non-fibrillar phosphorylated tau complexes in cortical homogenates from the brains of patients with AD effectively, at a concentration that was 1000-fold lower than for control homogenates [135]; thus, clinically safe doses of EC-102 are a possibility.
Large tau oligomers and fibrils are degraded through macroautophagy, a mechanism that can be induced by the drug sirolimus (rapamycin). Treatment with sirolimus reduced both the cellular levels of insoluble tau and the associated toxicity in Drosophila expressing wild-type or mutant forms of tau [139]. Unfortunately, because of the unintentional effects of sirolimus on mTOR signaling networks, this drug is not considered safe for the treatment of AD, despite the benefits yielded with treatment of tau-mutated Drosophila [140]. In addition, the use of sirolimus as an anti-tau agent in patients with AD is further restricted because this compound induces immunosuppression [111]. However, the inhibition of an mTOR-independent target (ie, inositol monophosphatase) by treatment with lithium chloride caused the upregulation of autophagy and beneficial changes in the levels of aggregated tau (in addition to the inhibition of GSK3, as described in the Inhibition of hyperphosphorylation section) [111]. Therefore, although much has yet to be elucidated with respect to the modulation of macroautophagy, the targeting of this process for the treatment of AD remains a possibility.
Attenuating the permeability of the mitochondrial permeability transition pore
Although Aβ-senile plaques and hyperphosphorylated tau aggregates are the most studied facets of AD, other pathological phenomena associated with this disease merit analysis. The mitochondrial permeability transition pore (mPTP) is a transmembrane protein complex that regulates the permeability of the inner mitochondrial membrane that is necessary for competent ETC function and ATP synthesis. Notably, in AD, mPTP breach occurs because of Aβ-induced stress [141], eliciting a collapse in the mitochondrial membrane potential (Δϕm) and amplifying the apoptotic mechanisms by the release of apoptogenic proteins from the inner-membrane space [142–144]. The antihistamine drug latrepirdine (Dimebon; Medivation Inc/Pfizer Inc) is an agent that has been demonstrated to block the opening of the mPTP effectively, and to protect against Aβ-induced cellular dysfunction and death [145–148]. In two trials in patients with mild-to-moderate AD, latrepirdine resulted in significant improvement in cognition (Mini Mental State Examination [MMSE] and ADAS-cog), function (Alzheimer's Disease Cooperative Study Activities of Daily Living Inventory [ADCS-ADL]) and behavior (Neuropsychiatric Inventory) scores [145,149]. Moreover, few adverse side effects were observed in both trials, thereby confirming the safety of latrepirdine [145,149]; the use of latrepirdine in Russia as an antihistamine for more than two decades is a further testament to the safety of this agent. The results of the latrepirdine phase III trial, which are expected in early 2010, are anticipated with interest.
Inhibiting glutamate-mediated excitotoxicity
Glutamate-mediated excitotoxicity is thought to contribute to the selective neurodegeneration observed in AD [93]. The inhibition of glutamate-mediated excitotoxicity has provided some therapeutic benefits in patients with mild-to-moderate AD. In particular, the NMDA receptor antagonist memantine is the only treatment available for AD that targets neurodegeneration [93,150]. Glutamate-mediated excitotoxicity is thought to be the result of the overactivation of the NMDA glutamate receptors, caused by an impaired uptake of excitatory amino acids [93]. Consequently, an interruption to this excitotoxic pathway prevents cells from succumbing to excitotoxic death, while enabling normal cognitive activation and memory formation. Recent studies have implicated memantine in the prevention of APP and tau expression via the inhibition of a translation initiation mechanism that is mediated by the internal ribosome entry site [150]. While the actions of memantine are beneficial, regardless of its mechanism of action, the overall effectiveness of this agent in delaying the progression of AD is modest [15,151,152].
Reducing luteinizing hormone concentrations
Considerable evidence exists for the role of an altered hormonal balance in patients with AD, suggesting that treatments that attenuate the altered hormonal homeostasis in an individual with the disease may be beneficial. Women have an approximately 2-fold increased risk of developing AD compared with men [153]. The increased incidence of AD in women has been attributed to menopause or age-related changes in the hypothalamic-pituitary-gonadal axis hormone levels, such as declines in sex steroids [154]. In Down's syndrome (DS), in which affected patients develop AD at a relatively early age [155], the gonadotropin luteinizing hormone (LH) is considered to have a role in the development of AD. Specifically, men with DS have an increased prevalence of AD, and hormonal investigations indicate that men with DS have higher levels of circulating LH compared with women with DS, while the reverse is true in healthy individuals [156]. Moreover, LH is the only hormonal factor that explains the gender predisposition for AD in both the healthy population and in those individuals with DS. Consequently, studies on LH and LH-based therapies have yielded exciting and promising results.
Individuals with AD have a 2-fold increase in serum LH concentrations, as compared with age-matched controls [157,158], and also have increased levels of LH in the hippocampus [155,156,159]. Furthermore, LH was demonstrated to drive APP toward the amyloidogenic pathway in vitro [160]. The gonadotropin-releasing hormone (GnRH) analog and agonist leuprolide acetate specifically reduces LH concentrations through the downregulation of GnRH receptors in the anterior pituitary [93]. In APP-mutant Tg mice (Tg2576) [156,160] and humans, the agent was demonstrated to reduce Aβ deposition [156] and to modulate cognitive performance, respectively (NCT00076440). Phase II clinical trials with leuprolide acetate indicated that high doses of the agent stabilized cognitive decline (as assessed by ADAS-Cog and ADCS-CGIC [ADCS Clinical Global Impression of Change] scores) and daily living activities (as assessed by the ADCS-ADL scale) (NCT00063310).
Symptomatic treatments for Alzheimer's disease
As researchers continue to investigate drugs that may prevent or reverse AD, the current burden of this disease on society demands some form of treatment strategy for the individuals affected. Although current treatments do not provide a long-term reduction of AD pathology or the reversal of disease progression, these therapies nonetheless postpone the inevitable decline in cognition by attenuating the epiphenomena of AD. Currently, the most widely used of these symptomatic treatments are cholinesterase inhibitors.
Cholinesterase inhibitors inhibit the enzymatic degradation of the neurotransmitter ACh by blocking the activity of AChE [161,162]. Because cholinergic neurotransmission is lacking in patients with AD, most notably in the basal forebrain [163], an increase in the synaptic levels of ACh enhances cognitive function [93]. Specifically, 50% of patients that undergo AChE inhibitor therapy report cognitive stability for 1 year; in addition, the beneficial effects of AChE inhibitor therapy can be maintained for up to 36 months [164–166]. However, although AChE inhibitor therapy is beneficial, a pathological reversal of, or cure for, AD is not achieved, and these treatments therefore represent only a short-term therapeutic approach. The positive impact on cognition, however, is apparent, and is clinically valuable.
Conclusion
As outlined in this review, much progress has been made in the search for an effective treatment for AD. Agents that potentially prevent or delay the progression of AD are being assessed in preclinical studies and clinical trials. Preventive measures (eg, antioxidants, mitochondrial antioxidants and mitochondrial dynamics protectors) will benefit an individual only prior to the onset of clinical dementia. After clinical dementia has been reached, the direct methods of preserving neuronal operational integrity will not fully impede the cascade of oxidative stress, mitochondrial decay, Aβ secretions, NFT accumulation and tau hyperphosphorylation. Disease-modifying remedies have been designed to treat mild-to-moderate AD. The development of drugs that are able to reduce and clear Aβ, to stabilize the tau and microtubule proteins, to protect the mitochondrial membrane potential and to prevent excitotoxicity ideally will halt the cascade that leads to the development of AD. With the development of such drugs, the attenuation of AD epiphenomena will not be required, as the disease will be treated successfully. Ultimately, although the majority of therapeutics will require a long process of clinical development, the overall successes described in this review indicate that an effective prevention treatment or cure for AD is possible.
Acknowledgements
Research in the authors' laboratories is supported by the NIH (AG028679 awarded to Mark A Smith; AG031853 awarded to Xiongwei Zhu) and the Alzheimer's Association.
Abbreviations
- Aβ
β-Amyloid
- APP
Aβ precursor protein
- AD
Alzheimer's disease
- ADAS-cog
Alzheimer's Disease Assessment Scale Cognitive Subscale
- ADCS-ADL
Alzheimer's Disease Cooperative Study Activities of Daily Living Inventory
- BBB
blood-brain barrier
- ETC
electron transport chain
- NFT
neurofibrillary tangle
- mtDNA
mitochondrial DNA
- OXPHOS
oxidative phosphorylation
- ROS
reactive oxidative species
- Tg
transgenic
Footnotes
Disclosure
Mark A Smith is or has been previously a paid consultant for, owns equity or stock options in, and/or receives grant funding from Advana Science Inc, Anavex Life Sciences Corp, Lytmos Group Inc, Medivation, Neurotez Inc, Olson Research Group, Panacea Pharmaceuticals Inc, Pfizer and Voyager Pharmaceutical Corp.
References
• of special interest
- 1.Mancuso M, Orsucci D, Siciliano G, Murri L. Mitochondria, mitochondrial DNA and Alzheimer's disease. What comes first? Curr Alzheimer Res. 2008;5(5):457–468. doi: 10.2174/156720508785908946. [DOI] [PubMed] [Google Scholar]
- 2.Zhu X, Lee HG, Perry G, Smith MA. Alzheimer disease, the two-hit hypothesis: An update. Biochim Biophys Acta. 2007;1772(4):494–502. doi: 10.1016/j.bbadis.2006.10.014. [DOI] [PubMed] [Google Scholar]
- 3.Selkoe DJ. Toward a comprehensive theory for Alzheimer's disease. Hypothesis: Alzheimer's disease is caused by the cerebral accumulation and cytotoxicity of amyloid β-protein. Ann N Y Acad Sci. 2000;924:17–25. doi: 10.1111/j.1749-6632.2000.tb05554.x. [DOI] [PubMed] [Google Scholar]
- 4.Smith MA, Nunomura A, Zhu X, Takeda A, Perry G. Metabolic, metallic, and mitotic sources of oxidative stress in Alzheimer disease. Antioxid Redox Signal. 2000;2(3):413–420. doi: 10.1089/15230860050192198. [DOI] [PubMed] [Google Scholar]
- 5.Atamna H, Frey WH., 2nd Mechanisms of mitochondrial dysfunction and energy deficiency in Alzheimer's disease. Mitochondrion. 2007;7(5):297–310. doi: 10.1016/j.mito.2007.06.001. [DOI] [PubMed] [Google Scholar]
- 6.Iqbal K, Zaidi T, Thompson CH, Merz PA, Wisniewski HM. Alzheimer paired helical filaments: Bulk isolation, solubility, and protein composition. Acta Neuropathol. 1984;62(3):167–177. doi: 10.1007/BF00691849. [DOI] [PubMed] [Google Scholar]
- 7.Grundke-Iqbal I, Iqbal K, Tung YC, Quinlan M, Wisniewski HM, Binder LI. Abnormal phosphorylation of the microtubule-associated protein tau (tau) in Alzheimer cytoskeletal pathology. Proc Natl Acad Sci USA. 1986;83(13):4913–4917. doi: 10.1073/pnas.83.13.4913. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Castellani RJ, Lee HG, Zhu X, Nunomura A, Perry G, Smith MA. Neuropathology of Alzheimer disease: Pathognomonic but not pathogenic. Acta Neuropathol. 2006;111(6):503–509. doi: 10.1007/s00401-006-0071-y. [DOI] [PubMed] [Google Scholar]
- 9. Nunomura A, Perry G, Aliev G, Hirai K, Takeda A, Balraj EK, Jones PK, Ghanbari H, Wataya T, Shimohama S, Chiba S, et al. Oxidative damage is the earliest event in Alzheimer disease. J Neuropathol Exp Neurol. 2001;60(8):759–767. doi: 10.1093/jnen/60.8.759.. • Oxidatively damaged RNA in AD is highest early in the course of disease, at a point when amyloid deposition is at low levels. Oxidatively damaged RNA provides a good target for early intervention.
- 10.Morsch R, Simon W, Coleman PD. Neurons may live for decades with neurofibrillary tangles. J Neuropathol Exp Neurol. 1999;58(2):188–197. doi: 10.1097/00005072-199902000-00008. [DOI] [PubMed] [Google Scholar]
- 11.Zhu X, Castellani RJ, Takeda A, Nunomura A, Atwood CS, Perry G, Smith MA. Differential activation of neuronal ERK, NK/SAPK, p38 in Alzheimer disease: The 'two hit' hypothesis. Mech Ageing Dev. 2001;123(1):39–46. doi: 10.1016/s0047-6374(01)00342-6. [DOI] [PubMed] [Google Scholar]
- 12.Wang X, Su B, Zheng L, Perry G, Smith MA, Zhu X. The role of abnormal mitochondrial dynamics in the pathogenesis of Alzheimer's disease. J Neurochem. 2009;109 Suppl 1:153–159. doi: 10.1111/j.1471-4159.2009.05867.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Zhu X, Raina AK, Perry G, Smith MA. Alzheimer's disease: The two-hit hypothesis. Lancet Neurol. 2004;3(4):219–226. doi: 10.1016/S1474-4422(04)00707-0. [DOI] [PubMed] [Google Scholar]
- 14.Hardy J. The amyloid hypothesis for Alzheimer's disease: A critical reappraisal. J Neurochem. 2009;110(4):1129–1134. doi: 10.1111/j.1471-4159.2009.06181.x. [DOI] [PubMed] [Google Scholar]
- 15.Smith MA, Richey Harris PL, Sayre LM, Beckman JS, Perry G. Widespread peroxynitrite-mediated damage in Alzheimer's disease. J Neurosci. 1997;17(8):2653–2657. doi: 10.1523/JNEUROSCI.17-08-02653.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Kalaria RN. Microglia and Alzheimer's disease. Curr Opin Hematol. 1999;6(1):15–24. doi: 10.1097/00062752-199901000-00004. [DOI] [PubMed] [Google Scholar]
- 17.Braun JS, Sublett JE, Freyer D, Mitchell TJ, Cleveland JL, Tuomanen EI, Weber JR. Pneumococcal pneumolysin and H2O2 mediate brain cell apoptosis during meningitis. J Clin Invest. 2002;109(1):19–27. doi: 10.1172/JCI12035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Eastman CL, Guilarte TR. The role of hydrogen peroxide in the in vitro cytotoxicity of 3-hydroxykynurenine. Neurochem Res. 1990;15(11):1101–1107. doi: 10.1007/BF01101711. [DOI] [PubMed] [Google Scholar]
- 19.Klivenyi P, Toldi J, Vecsei L. Kynurenines in neurodegenerative disorders: Therapeutic consideration. Adv Exp Med Biol. 2004;541:169–183. doi: 10.1007/978-1-4419-8969-7_10. [DOI] [PubMed] [Google Scholar]
- 20.Schwarcz R, Köhler C. Differential vulnerability of central neurons of the rat to quinolinic acid. Neurosci Lett. 1983;38(1):85–90. doi: 10.1016/0304-3940(83)90115-5. [DOI] [PubMed] [Google Scholar]
- 21.Schwarcz R, Foster AC, French ED, Whetsell WO, Jr, Köhler C. Excitotoxic models for neurodegenerative disorders. Life Sci. 1984;35(1):19–32. doi: 10.1016/0024-3205(84)90148-6. [DOI] [PubMed] [Google Scholar]
- 22.Stone TW, Perkins MN. Quinolinic acid: A potent endogenous excitant at amino acid receptors in CNS. Eur J Pharmacol. 1981;72(4):411–412. doi: 10.1016/0014-2999(81)90587-2. [DOI] [PubMed] [Google Scholar]
- 23.Guillemin GJ, Brew BJ. Implications of the kynurenine pathway and quinolinic acid in Alzheimer's disease. Redox Rep. 2002;7(4):199–206. doi: 10.1179/135100002125000550. [DOI] [PubMed] [Google Scholar]
- 24.Hempen B, Brion JP. Reduction of acetylated α-tubulin immunoreactivity in neurofibrillary tangle-bearing neurons in Alzheimer's disease. J Neuropathol Exp Neurol. 1996;55(9):964–972. doi: 10.1097/00005072-199609000-00003. [DOI] [PubMed] [Google Scholar]
- 25.Bendiske J, Caba E, Brown QB, Bahr BA. Intracellular deposition microtubule destabilization transport failure: An 'early' pathogenic cascade leading to synaptic decline. J Neuropathol Exp Neurol. 2002;61(7):640–650. doi: 10.1093/jnen/61.7.640. [DOI] [PubMed] [Google Scholar]
- 26.Su B, Wang X, Zheng L, Perry G, Smith MA, Zhu X. Abnormal mitochondrial dynamics and neurodegenerative diseases. Biochim Biophys Acta. 2010;1802(1):135–142. doi: 10.1016/j.bbadis.2009.09.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Castellani RJ, Harris PL, Sayre LM, Fujii J, Taniguchi N, Vitek MP, Founds H, Atwood CS, Perry G, Smith MA. Active glycation in neurofibrillary pathology of Alzheimer disease: Nε-(carboxymethyl) lysine and hexitol-lysine. Free Radic Biol Med. 2001;31(2):175–180. doi: 10.1016/s0891-5849(01)00570-6. [DOI] [PubMed] [Google Scholar]
- 28. Bonda DJ, Wang X, Perry G, Smith MA, Zhu X. Mitochondrial dynamics in Alzheimer disease: Opportunities for future treatment strategies. Drugs Aging. 2010;27:1–12. doi: 10.2165/11532140-000000000-00000.. • As both sources of ROS and sites of degeneration in AD, mitochondria function and structure must be protected to prevent neurodegeneration.
- 29.Petersen RB, Nunomura A, Lee HG, Casadesus G, Perry G, Smith MA, Zhu X. Signal transduction cascades associated with oxidative stress in Alzheimer's disease. J Alzheimers Dis. 2007;11(2):143–152. doi: 10.3233/jad-2007-11202. [DOI] [PubMed] [Google Scholar]
- 30.Perrig WJ, Perrig P, Stähelin HB. The relation between antioxidants and memory performance in the old and very old. J Am Geriatr Soc. 1997;45(6):718–724. doi: 10.1111/j.1532-5415.1997.tb01476.x. [DOI] [PubMed] [Google Scholar]
- 31.Zandi PP, Anthony JC, Khachaturian AS, Stone SV, Gustafson D, Tschanz JT, Norton MC, Welsh-Bohmer KA, Breitner JC. Reduced risk of Alzheimer disease in users of antioxidant vitamin supplements: The Cache County Study. Arch Neurol. 2004;61(1):82–88. doi: 10.1001/archneur.61.1.82. [DOI] [PubMed] [Google Scholar]
- 32.Boothby LA, Doering PL. Vitamin C and vitamin E for Alzheimer's disease. Ann Pharmacother. 2005;39(12):2073–2080. doi: 10.1345/aph.1E495. [DOI] [PubMed] [Google Scholar]
- 33.Sano M, Ernesto C, Thomas RG, Klauber MR, Schafer K, Grundman M, Woodbury P, Growdon J, Cotman CW, Pfeiffer E, Schneider LS, et al. A controlled trial of selegiline, α-tocopherol, or both as treatment for Alzheimer's disease. The Alzheimer's Disease Cooperative Study. N Engl J Med. 1997;336(17):1245–1247. doi: 10.1056/NEJM199704243361704. [DOI] [PubMed] [Google Scholar]
- 34.Lee YJ, Jeong SY, Karbowski M, Smith CL, Youle RJ. Roles of the mammalian mitochondrial fission and fusion mediators Fis1, Drp1, and Opa1 in apoptosis. Mol Biol Cell. 2004;15(11):5001–5011. doi: 10.1091/mbc.E04-04-0294. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Zhu X, Perry G, Moreira PI, Aliev G, Cash AD, Hirai K, Smith VA. Mitochondrial abnormalities and oxidative imbalance in Alzheimer disease. J Alzheimers Dis. 2006;9(2):147–153. doi: 10.3233/jad-2006-9207. [DOI] [PubMed] [Google Scholar]
- 36.Aliev G, Smith MA, Obrenovich ME, de la Torre JC, Perry G. Role of vascular hypoperfusion-induced oxidative stress and mitochondria failure in the pathogenesis of Alzheimer disease. Neurotox Res. 2003;5(7):491–504. doi: 10.1007/BF03033159. [DOI] [PubMed] [Google Scholar]
- 37.Ogawa O, Zhu X, Perry G, Smith MA. Mitochondrial abnormalities and oxidative imbalance in neurodegenerative disease. Sci Aging Knowledge Environ. 2002;2002(41):pe16. doi: 10.1126/sageke.2002.41.pe16. [DOI] [PubMed] [Google Scholar]
- 38.Lee J, Boo JH, Ryu H. The failure of mitochondria leads to neurodegeneration: Do mitochondria need a jump start? Adv Drug Deliv Rev. 2009;61(14):1316–1323. doi: 10.1016/j.addr.2009.07.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Beal MF. Mitochondrial dysfunction and oxidative damage in Alzheimer's and Parkinson's diseases and coenzyme Q10 as a potential treatment. J Bioenerg Biomembr. 2004;36(4):381–386. doi: 10.1023/B:JOBB.0000041772.74810.92. [DOI] [PubMed] [Google Scholar]
- 40.Wadsworth TL, Bishop JA, Pappu AS, Woltjer RL, Quinn JF. Evaluation of coenzyme Q as an antioxidant strategy for Alzheimer's disease. J Alzheimers Dis. 2008;14(2):225–234. doi: 10.3233/jad-2008-14210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Lass A, Forster MJ, Sohal RS. Effects of coenzyme Q10 and α-tocopherol administration on their tissue levels in the mouse: Elevation of mitochondrial α-tocopherol by coenzyme Q10. Free Radic Biol Med. 1999;26(11–12):1375–1382. doi: 10.1016/s0891-5849(98)00330-x. [DOI] [PubMed] [Google Scholar]
- 42.Kwong LK, Kamzalov S, Rebrin I, Bayne AC, Jana CK, Morris P, Forster MJ, Sohal RS. Effects of coenzyme Q10 administration on its tissue concentrations, mitochondrial oxidant generation, and oxidative stress in the rat. Free Radic Biol Med. 2002;33(5):627–638. doi: 10.1016/s0891-5849(02)00916-4. [DOI] [PubMed] [Google Scholar]
- 43.Murphy MP. Development of lipophilic cations as therapies for disorders e. mitochondrial dysfunction. Expert Opin Biol Ther. 2001;1(5):753–764. doi: 10.1517/14712598.1.5.753. [DOI] [PubMed] [Google Scholar]
- 44.Smith RA, Kelso GF, James AM, Murphy MP. Targeting coenzyme Q derivatives to mitochondria. Methods Enzymol. 2004;382:45–67. doi: 10.1016/S0076-6879(04)82003-2. [DOI] [PubMed] [Google Scholar]
- 45.Lu C, Zhang D, Whiteman M, Armstrong JS. Is antioxidant potential of the mitochondrial targeted ubiquinone derivative MitoQ conserved in cells lacking mtDNA? Antioxid Redox Signal. 2008;10(3):651–660. doi: 10.1089/ars.2007.1865. [DOI] [PubMed] [Google Scholar]
- 46.Murphy MP, Smith RA. Targeting antioxidants to mitochondria by conjugation to lipophilic cations. Annu Rev Pharmacol Toxicol. 2007;47:629–656. doi: 10.1146/annurev.pharmtox.47.120505.105110. [DOI] [PubMed] [Google Scholar]
- 47.Tauskela JS. MitoQ - A mitochondria-targeted antioxidant. IDrugs. 2007;10(6):399–412. [PubMed] [Google Scholar]
- 48.Siedlak SL, Casadesus G, Webber KM, Pappolla MA, Atwood CS, Smith MA, Perry G. Chronic antioxidant therapy reduces oxidative stress in a mouse model of Alzheimer's disease. Free Radic Res. 2009;43(2):156–164. doi: 10.1080/10715760802644694. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Liu J, Head E, Gharib AM, Yuan W, Ingersoll RT, Hagen TM, Cotman CW, Ames BN. Memory loss in old rats is associated with brain mitochondrial decay and RNA/DNA oxidation: Partial reversal by feeding acetyl-l-carnitine and/or R-α-lipoic acid. Proc Natl Acad Sci USA. 2002;99(4):2356–2361. doi: 10.1073/pnas.261709299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Liu J, Atamna H, Kuratsune H, Ames BN. Delaying brain mitochondrial decay and aging with mitochondrial antioxidants and metabolites. Ann N Y Acad Sci. 2002;959:133–166. doi: 10.1111/j.1749-6632.2002.tb02090.x. [DOI] [PubMed] [Google Scholar]
- 51.Long J, Gao F, Tong L, Cotman CW, Ames BN, Liu J. Mitochondrial decay in the brains of old rats: Ameliorating effect of α-lipoic acid and acetyl-l-carnitine. Neurochem Res. 2009;34(4):755–763. doi: 10.1007/s11064-008-9850-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Aliev G, Liu J, Shenk JC, Fischbach K, Pacheco GJ, Chen SG, Obrenovich ME, Ward WF, Richardson AG, Smith MA, Gasimov E, et al. Neuronal mitochondrial amelioration by feeding acetyl-l-carnitine and lipoic acid to aged rats. J Cell Mol Med. 2009;13(2):320–333. doi: 10.1111/j.1582-4934.2008.00324.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Liu J, Killilea DW, Ames BN. Age-associated mitochondrial oxidative decay: Improvement of carnitine acetyltransferase substrate-binding affinity and activity in brain by feeding old rats acetyl-l- carnitine and/or R-α-lipoic acid. Proc Natl Acad Sci USA. 2002;99(4):1876–1881. doi: 10.1073/pnas.261709098. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Milgram NW, Araujo JA, Hagen TM, Treadwell BV, Ames BN. Acetyl-l-carnitine and α-lipoic acid supplementation of aged beagle dogs improves learning in two landmark discrimination tests. FASEB J. 2007;21(13):3756–3762. doi: 10.1096/fj.07-8531com. [DOI] [PubMed] [Google Scholar]
- 55.Ames BN, Liu J. Delaying the mitochondrial decay of aging with acetylcarnitine. Ann N Y Acad Sci. 2004;1033:108–116. doi: 10.1196/annals.1320.010. [DOI] [PubMed] [Google Scholar]
- 56.Chan DC. Mitochondrial fusion and fission in mammals. Annu Rev Cell Dev Biol. 2006;22:79–99. doi: 10.1146/annurev.cellbio.22.010305.104638. [DOI] [PubMed] [Google Scholar]
- 57.Chan DC. Mitochondria: Dynamic organelles in disease, aging, and development. Cell. 2006;125(7):1241–1252. doi: 10.1016/j.cell.2006.06.010. [DOI] [PubMed] [Google Scholar]
- 58.Chen H, Chomyn A, Chan DC. Disruption of fusion results in mitochondrial heterogeneity and dysfunction. J Biol Chem. 2005;280(28):26185–26192. doi: 10.1074/jbc.M503062200. [DOI] [PubMed] [Google Scholar]
- 59.Chen H, McCaffery JM, Chan DC. Mitochondrial fusion protects against neurodegeneration in the cerebellum. Cell. 2007;130(3):548–562. doi: 10.1016/j.cell.2007.06.026. [DOI] [PubMed] [Google Scholar]
- 60.Twig G, Elorza A, Molina AJ, Mohamed H, Wikstrom JD, Walzer G, Stiles L, Haigh SE, Katz S, Las G, Alroy J, et al. Fission and selective fusion govern mitochondrial segregation and elimination by autophagy. EMBO J. 2008;27(2):433–446. doi: 10.1038/sj.emboj.7601963. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Hirai K, Aliev G, Nunomura A, Fujioka H, Russell RL, Atwood CS, Johnson AB, Kress Y, Vinters HV, Tabaton M, Shimohama S, et al. Mitochondrial abnormalities in Alzheimer's disease. J Neurosci. 2001;21(9):3017–3023. doi: 10.1523/JNEUROSCI.21-09-03017.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Wang X, Su B, Lee HG, Li X, Perry G, Smith MA, Zhu X. Impaired balance of mitochondrial fission and fusion in Alzheimer's disease. J Neurosci. 2009;29(28):9090–9103. doi: 10.1523/JNEUROSCI.1357-09.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Wang X, Su B, Siedlak SL, Moreira PI, Fujioka H, Wang Y, Casadesus G, Zhu X. Amyloid-β overproduction causes abnormal mitochondrial dynamics via differential modulation of mitochondrial fission/fusion proteins. Proc Natl Acad Sci USA. 2008;105(49):19318–19323. doi: 10.1073/pnas.0804871105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Wang X, Su B, Fujioka H, Zhu X. Dynamin-like protein 1 reduction underlies mitochondrial morphology and distribution abnormalities in fibroblasts from sporadic Alzheimer's disease patients. Am J Pathol. 2008;173(2):470–482. doi: 10.2353/ajpath.2008.071208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Hayashi T, Shishido N, Nakayama K, Nunomura A, Smith MA, Perry G, Nakamura M. Lipid peroxidation and 4-hydroxy-2-nonenal formation by copper ion bound to amyloid-β peptide. Free Radic Biol Med. 2007;43(11):1552–1559. doi: 10.1016/j.freeradbiomed.2007.08.013. [DOI] [PubMed] [Google Scholar]
- 66.Nakamura M, Shishido N, Nunomura A, Smith MA, Perry G, Hayashi Y, Nakayama K, Hayashi T. Three histidine residues of amyloid-β peptide control the redox activity of copper and iron. Biochemistry. 2007;46(44):12737–12743. doi: 10.1021/bi701079z. [DOI] [PubMed] [Google Scholar]
- 67.Cuajungco MP, Goldstein LE, Nunomura A, Smith MA, Lim JT, Atwood CS, Huang X, Farrag YW, Perry G, Bush AI. Evidence that the β-amyloid plaques of Alzheimer's disease represent the redox-silencing and entombment of Aβ by zinc. J Biol Chem. 2000;275(26):19439–19442. doi: 10.1074/jbc.C000165200. [DOI] [PubMed] [Google Scholar]
- 68.Kontush A, Berndt C, Weber W, Akopyan V, Arlt S, Schippling S, Beisiegel U. Amyloid-β is an antioxidant for lipoproteins in cerebrospinal fluid and plasma. Free Radic Biol Med. 2001;30(1):119–128. doi: 10.1016/s0891-5849(00)00458-5. [DOI] [PubMed] [Google Scholar]
- 69.Atwood CS, Moir RD, Huang X, Scarpa RC, Bacarra NM, Romano DM, Hartshorn MA, Tanzi RE, Bush AI. Dramatic aggregation of Alzheimer Aβ by CuII is induced by conditions representing physiological acidosis. J Biol Chem. 1998;273(21):12817–12826. doi: 10.1074/jbc.273.21.12817. [DOI] [PubMed] [Google Scholar]
- 70.Atwood CS, Smith MA, Martins RN, Tanzi RE, Roher AE, Bush AI, Perry G. Neuroinflammatory environments promote amyloid-β deposition and posttranslational modification. In: Wood PL, editor. Neuroinflammation: Mechanisms and Management. Totowa, NJ, USA: Humana Press Inc; 2003. pp. 249–266. [Google Scholar]
- 71.Rottkamp CA, Raina AK, Zhu X, Gaier E, Bush AI, Atwood CS, Chevion M, Perry G, Smith MA. Redox-active iron mediates amyloid-β toxicity. Free Radic Biol Med. 2001;30(4):447–450. doi: 10.1016/s0891-5849(00)00494-9. [DOI] [PubMed] [Google Scholar]
- 72.Masters CL, Simms G, Weinman NA, Multhaup G, McDonald BL, Beyreuther K. Amyloid plaque core protein in Alzheimer disease and Down syndrome. Proc Natl Acad Sci USA. 1985;82(12):4245–4249. doi: 10.1073/pnas.82.12.4245. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Smith MA, Zhu X, Tabaton M, Liu G, McKeel DW, Jr, Cohen ML, Wang X, Siedlak SL, Dwyer BE, Hayashi T, Nakamura M, et al. Increased iron and free radical generation in preclinical Alzheimer disease and mild cognitive impairment. J Alzheimers Dis. 2010;19(1):363–372. doi: 10.3233/JAD-2010-1239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Honda K, Casadesus G, Petersen RB, Perry G, Smith MA. Oxidative stress and redox-active iron in Alzheimer's disease. Ann N Y Acad Sci. 2004;1012:179–182. doi: 10.1196/annals.1306.015. [DOI] [PubMed] [Google Scholar]
- 75.Atwood CS, Scarpa RC, Huang X, Moir RD, Jones WD, Fairlie DP, Tanzi RE, Bush AI. Characterization of copper interactions with Alzheimer amyloid β peptides: Identification of an attomolar-affinity copper binding site on amyloid β1–42. J Neurochem. 2000;75(3):1219–1233. doi: 10.1046/j.1471-4159.2000.0751219.x. [DOI] [PubMed] [Google Scholar]
- 76.Bush AI, Pettingell WH, Multhaup G, d Paradis M, Vonsattel JP, Gusella JF, Beyreuther K, Masters CL, Tanzi RE. Rapid induction of Alzheimer Aβ amyloid formation by zinc. Science. 1994;265(5177):1464–1467. doi: 10.1126/science.8073293. [DOI] [PubMed] [Google Scholar]
- 77.Exley C. Aluminium and iron, but neither copper nor zinc, are key to the precipitation of β-sheets of Aβ42 in senile plaque cores in Alzheimer's disease. J Alzheimers Dis. 2006;10(2–3):173–177. doi: 10.3233/jad-2006-102-305. [DOI] [PubMed] [Google Scholar]
- 78.Jobling MF, Huang X, Stewart LR, Barnham KJ, Curtain C, Volitakis I, Perugini M, White AR, Cherny RA, Masters CL, Barrow CJ, Collins SJ, Bush AI, Cappai R. Copper and zinc binding modulates the aggregation and neurotoxic properties of the prion peptide PrP106–126. Biochemistry. 2001;40(27):8073–8084. doi: 10.1021/bi0029088. [DOI] [PubMed] [Google Scholar]
- 79.Crapper McLachlan DR, Dalton AJ, Kruck TP, Bell MY, Smith WL, Kalow W, Andrews DF. Intramuscular desferrioxamine in patients with Alzheimer's disease. Lancet. 1991;337(8753):1304–1308. doi: 10.1016/0140-6736(91)92978-b. [DOI] [PubMed] [Google Scholar]
- 80.Regland B, Lehmann W, Abedini I, Blennow K, Jonsson M, Karlsson I, Sjogren M, Wallin A, Xilinas M, Gottfries CG. Treatment of Alzheimer's disease with clioquinol. Dement Geriatr Cogn Disord. 2001;12(6):408–414. doi: 10.1159/000051288. [DOI] [PubMed] [Google Scholar]
- 81.Ritchie CW, Bush AI, Mackinnon A, Macfarlane S, Mastwyk M, MacGregor L, Kiers L, Cherny R, Li QX, Tammer A, Carrington D, et al. Metal-protein attenuation with iodochlorhydroxyquin (clioquinol) targeting Aβ amyloid deposition and toxicity in Alzheimer disease: A pilot phase 2 clinical trial. Arch Neurol. 2003;60(12):1685–1691. doi: 10.1001/archneur.60.12.1685. [DOI] [PubMed] [Google Scholar]
- 82.Huang X, Atwood CS, Hartshorn MA, Multhaup G, Goldstein LE, Scarpa RC, Cuajungco MP, Gray DN, Lim J, Moir RD, Tanzi RE, et al. The Aβ peptide of Alzheimer's disease directly produces hydrogen peroxide through metal ion reduction. Biochemistry. 1999;38(24):7609–7616. doi: 10.1021/bi990438f. [DOI] [PubMed] [Google Scholar]
- 83.Opazo C, Huang X, Cherny RA, Moir RD, Roher AE, White AR, Cappai R, Masters CL, Tanzi RE, Inestrosa NC, Bush AI. Metalloenzyme-like activity of Alzheimer's disease β-amyloid. Cu-dependent catalytic conversion of dopamine, cholesterol, and biological reducing agents to neurotoxic H2O2. J Biol Chem. 2002;277(43):40302–40308. doi: 10.1074/jbc.M206428200. [DOI] [PubMed] [Google Scholar]
- 84.Liu G, Men P, Harris PL, Rolston RK, Perry G, Smith MA. Nanoparticle iron chelators: A new therapeutic approach in Alzheimer disease and other neurologic disorders associated with trace metal imbalance. Neurosci Lett. 2006;406(3):189–193. doi: 10.1016/j.neulet.2006.07.020. [DOI] [PubMed] [Google Scholar]
- 85.Schubert D, Chevion M. The role of iron in β amyloid toxicity. Biochem Biophys Res Commun. 1995;216(2):702–707. doi: 10.1006/bbrc.1995.2678. [DOI] [PubMed] [Google Scholar]
- 86.Liu G, Men P, Kudo W, Perry G, Smith MA. Nanoparticle-chelator conjugates as inhibitors of amyloid-β aggregation and neurotoxicity: A novel therapeutic approach for Alzheimer disease. Neurosci Lett. 2009;455(3):187–190. doi: 10.1016/j.neulet.2009.03.064. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Olcese JM, Cao C, Mori T, Mamcarz MB, Maxwell A, Runfeldt MJ, Wang L, Zhang C, Lin X, Zhang G, Arendash GW. Protection against cognitive deficits and markers of neurodegeneration by long-term oral administration of melatonin in a transgenic model of Alzheimer disease. J Pineal Res. 2009;47(1):82–96. doi: 10.1111/j.1600-079X.2009.00692.x. [DOI] [PubMed] [Google Scholar]
- 88.Pappolla M, Bozner P, Soto C, Shao H, Robakis NK, Zagorski M, Frangione B, Ghiso J. Inhibition of Alzheimer β-fibrillogenesis by melatonin. J Biol Chem. 1998;273(13):7185–7188. doi: 10.1074/jbc.273.13.7185. [DOI] [PubMed] [Google Scholar]
- 89.Poeggeler B, Miravalle L, Zagorski MG, Wisniewski T, Chyan YJ, Zhang Y, Shao H, Bryant-Thomas T, Vidal R, Frangione B, Ghiso J, et al. Melatonin reverses the profibrillogenic activity of apolipoprotein E4 on the Alzheimer amyloid Aβ peptide. Biochemistry. 2001;40(49):14995–15001. doi: 10.1021/bi0114269. [DOI] [PubMed] [Google Scholar]
- 90.Fraser PE, Nguyen JT, Surewicz WK, Kirschner DA. pH-dependent structural transitions of Alzheimer amyloid peptides. Biophys J. 1991;60(5):1190–1201. doi: 10.1016/S0006-3495(91)82154-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Sabbagh MN. Drug development for Alzheimer's disease: Where are we now and where are we headed? Am J Geriatr Pharmacother. 2009;7(3):167–185. doi: 10.1016/j.amjopharm.2009.06.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.DaSilva KA, Brown ME, Cousins JE, Rappaport RV, Aubert I, Westaway D, McLaurin J. Scyllo-inositol (ELND005) ameliorates amyloid pathology in an aggressive mouse model of Alzheimer's disease. Alzheimer's & Dementia. 2009;5 Suppl 1:P425. [Google Scholar]
- 93.Marlatt MW, Webber KM, Moreira PI, Lee HG, Casadesus G, Honda K, Zhu X, Perry G, Smith MA. Therapeutic opportunities in Alzheimer disease: One for all or all for one? Curr Med Chem. 2005;12(10):1137–1147. doi: 10.2174/0929867053764644. [DOI] [PubMed] [Google Scholar]
- 94.Lundkvist J, Naslund J. γ-secretase: A complex target for Alzheimer's disease. Curr Opin Pharmacol. 2007;7(1):112–118. doi: 10.1016/j.coph.2006.10.002. [DOI] [PubMed] [Google Scholar]
- 95.Barten DM, Guss VL, Corsa JA, Loo A, Hansel SB, Zheng M, Munoz B, Srinivasan K, Wang B, Robertson BJ, Polson CT, et al. Dynamics of β-amyloid reductions in brain, cerebrospinal fluid, and plasma of β-amyloid precursor protein transgenic mice treated with a γ-secretase inhibitor. J Pharmacol Exp Ther. 2005;312(2):635–643. doi: 10.1124/jpet.104.075408. [DOI] [PubMed] [Google Scholar]
- 96.Wong GT, Manfra D, Poulet FM, Zhang Q, Josien H, Bara T, Engstrom L, Pinzon-Ortiz M, Fine JS, Lee HJ, Zhang L, et al. Chronic treatment with the γ-secretase inhibitor LY-411,575 inhibits β-amyloid peptide production and alters lymphopoiesis and intestinal cell differentiation. J Biol Chem. 2004;279(13):12876–12882. doi: 10.1074/jbc.M311652200. [DOI] [PubMed] [Google Scholar]
- 97.Neugroschl J, Sano M. An update on treatment and prevention strategies for Alzheimer's disease. Curr Neurol Neurosci Rep. 2009;9(5):368–376. doi: 10.1007/s11910-009-0054-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Yan R. CHI's Eighth Annual World Pharmaceutical Congress. Targeting Alzheimer's with novel therapeutics: Part 2. IDrugs. 2009;12(8):482–484. [PubMed] [Google Scholar]
- 99.Perry G, Nunomura A, Raina AK, Smith MA. Amyloid-β junkies. Lancet. 2000;355(9205):757. doi: 10.1016/S0140-6736(05)72173-5. [DOI] [PubMed] [Google Scholar]
- 100.Smith MA, Atwood CS, Joseph JA, Perry G. Predicting the failure of amyloid-β vaccine. Lancet. 2002;359(9320):1864–1865. doi: 10.1016/S0140-6736(02)08695-6. [DOI] [PubMed] [Google Scholar]
- 101.Grundman M, Black R. O3–04–05: Clinical trials of bapineuzumab, a β-amyloid-targeted immunotherapy in patients with mild to moderate Alzheimer's disease. Alzheimers Dement. 2008;4(4):T166. [Google Scholar]
- 102.Relkin NR, Szabo P, Adamiak B, Burgut T, Monthe C, Lent RW, Younkin S, Younkin L, Schiff R, Weksler ME. 18-Month study of intravenous immunoglobulin for treatment of mild Alzheimer disease. Neurobiol Aging. 2009;30(11):1728–1736. doi: 10.1016/j.neurobiolaging.2007.12.021. [DOI] [PubMed] [Google Scholar]
- 103.Fassbender K, Simons M, Bergmann C, Stroick M, Lutjohann D, Keller P, Runz H, Kuhl S, Bertsch T, von Bergmann K, Hennerici M, et al. Simvastatin strongly reduces levels of Alzheimer's disease β-amyloid peptides Aβ42 and Aβ40in vitro and in vivo. Proc Natl Acad Sci USA. 2001;98(10):5856–5861. doi: 10.1073/pnas.081620098. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Nunomura A, Castellani RJ, Lee HG, Moreira PI, Zhu X, Perry G, Smith MA. Neuropathology in Alzheimer's disease: Awaking from a hundred-year-old dream. Sci Aging Knowledge Environ. 2006;2006(8):e10. doi: 10.1126/sageke.2006.8.pe10. [DOI] [PubMed] [Google Scholar]
- 105.Perry G, Rizzuto N, Autilio-Gambetti L, Gambetti P. Paired helical filaments from Alzheimer disease patients contain cytoskeletal components. Proc Natl Acad Sci USA. 1985;82(11):3916–3920. doi: 10.1073/pnas.82.11.3916. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Ishihara T, Hong M, Zhang B, Nakagawa Y, Lee MK, Trojanowski JQ, Lee VM. Age-dependent emergence and progression of a tauopathy in transgenic mice overexpressing the shortest human tau isoform. Neuron. 1999;24(3):751–762. doi: 10.1016/s0896-6273(00)81127-7. [DOI] [PubMed] [Google Scholar]
- 107.Cash AD, Aliev G, Siedlak SL, Nunomura A, Fujioka H, Zhu X, Raina AK, Vinters HV, Tabaton M, Johnson AB, Paula-Barbosa M, et al. Microtubule reduction in Alzheimer's disease and aging is independent of tau filament formation. Am J Pathol. 2003;162(5):1623–1627. doi: 10.1016/s0002-9440(10)64296-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Zhang B, Maiti A, Shively S, Lakhani F, McDonald-Jones G, Bruce J, Lee EB, Xie SX, Joyce S, Li C, Toleikis PM, et al. Microtubule-binding drugs offset tau sequestration by stabilizing microtubules and reversing fast axonal transport deficits in a tauopathy model. Proc Natl Acad Sci USA. 2005;102(1):227–231. doi: 10.1073/pnas.0406361102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Ballatore C, Hyde E, Deiches RF, Lee VM, Trojanowski JQ, Huryn D, Smith AB., 3rd Paclitaxel C-10 carbamates: Potential candidates for the treatment of neurodegenerative tauopathies. Bioorg Med Chem Lett. 2007;17(13):3642–3646. doi: 10.1016/j.bmcl.2007.04.058. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Fellner S, Bauer B, Miller DS, Schaffrik M, Fankhanel M, Spruss T, Bernhardt G, Graeff C, Farber L, Gschaidmeier H, Buschauer A, et al. Transport of paclitaxel (Taxol) across the blood-brain barrier in vitro and in vivo. J Clin Invest. 2002;110(9):1309–1318. doi: 10.1172/JCI15451. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Brunden KR, Trojanowski JQ, Lee VM. Advances in tau-focused drug discovery for Alzheimer's disease and related tauopathies. Nat Rev Drug Discov. 2009;8(10):783–793. doi: 10.1038/nrd2959. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Radaideh SM, Sledge GW. Taxane vs taxane: Is the duel at an end? A commentary on a phase-III trial of doxorubicin and docetaxel versus doxorubicin and paclitaxel in metastatic breast cancer: Results of the ERASME 3 study. Breast Cancer Res Treat. 2008;111(2):203–208. doi: 10.1007/s10549-007-9776-4. [DOI] [PubMed] [Google Scholar]
- 113.Matsuoka Y, Gray AJ, Hirata-Fukae C, Minami SS, Waterhouse EG, Mattson MP, LaFerla FM, Gozes I, Aisen PS. Intranasal NAP administration reduces accumulation of amyloid peptide and tau hyperphosphorylation in a transgenic mouse model of Alzheimer's disease at early pathological stage. J Mol Neurosci. 2007;31(2):165–170. doi: 10.1385/jmn/31:02:165. [DOI] [PubMed] [Google Scholar]
- 114.Matsuoka Y, Jouroukhin Y, Gray AJ, Ma L, Hirata-Fukae C, Li HF, Feng L, Lecanu L, Walker BR, Planel E, Arancio O, et al. A neuronal microtubule-interacting agent, NAPVSIPQ, reduces tau pathology and enhances cognitive function in a mouse model of Alzheimer's disease. J Pharmacol Exp Ther. 2008;325(1):146–153. doi: 10.1124/jpet.107.130526. [DOI] [PubMed] [Google Scholar]
- 115.Lee HG, Perry G, Moreira PI, Garrett MR, Liu Q, Zhu X, Takeda A, Nunomura A, Smith MA. Tau phosphorylation in Alzheimer's disease: Pathogen or protector? Trends Mol Med. 2005;11(4):164–169. doi: 10.1016/j.molmed.2005.02.008. [DOI] [PubMed] [Google Scholar]
- 116.Hanger DP, Anderton BH, Noble W. Tau phosphorylation: The therapeutic challenge for neurodegenerative disease. Trends Mol Med. 2009;15(3):112–119. doi: 10.1016/j.molmed.2009.01.003. [DOI] [PubMed] [Google Scholar]
- 117.Sengupta A, Kabat J, Novak M, Wu Q, Grundke-Iqbal I, Iqbal K. Phosphorylation of tau at both Thr231 and Ser262 is required for maximal inhibition of its binding to microtubules. Arch Biochem Biophys. 1998;357(2):299–309. doi: 10.1006/abbi.1998.0813. [DOI] [PubMed] [Google Scholar]
- 118.Schneider A, Biernat J, von Bergen M, Mandelkow E, Mandelkow EM. Phosphorylation that detaches tau protein from microtubules (Ser262, Ser214) also protects it against aggregation into Alzheimer paired helical filaments. Biochemistry. 1999;38(12):3549–3558. doi: 10.1021/bi981874p. [DOI] [PubMed] [Google Scholar]
- 119.Medina M, Castro A. Glycogen synthase kinase-3 (GSK-3) inhibitors reach the clinic. Curr Opin Drug Discov Devel. 2008;11(4):533–543. [PubMed] [Google Scholar]
- 120.Imahori K, Uchida T. Physiology and pathology of tau protein kinases in relation to Alzheimer's disease. J Biochem. 1997;121(2):179–188. [PubMed] [Google Scholar]
- 121.Lucas JJ, Hernández F, Gómez-Ramos P, Morán MA, Hen R, Avila J. Spatial learning deficit in transgenic mice that conditionally over-express GSK-3β in the brain but do not form tau filaments. J Neurochem. 2002;83(6):1529–1533. doi: 10.1046/j.1471-4159.2002.01269.x. [DOI] [PubMed] [Google Scholar]
- 122. Hernandez F, Borrell J, Guaza C, Avila J, Lucas JJ. Spatial learning deficit in transgenic mice that conditionally over-express GSK-3b in the brain but do not form tau filaments. J Neurochem. 2002;83(6):1529–1533. doi: 10.1046/j.1471-4159.2002.01269.x.. • Lithium chloride reduced the accumulation of tau by inhibiting GSK-3, and represents a future treatment option for AD.
- 123.Noble W, Planel E, Zehr C, Olm V, Meyerson J, Suleman F, Gaynor K, Wang L, LaFrancois J, Feinstein B, Burns M, et al. Inhibition of glycogen synthase kinase-3 by lithium correlates with reduced tauopathy and degeneration in vivo. Proc Natl Acad Sci USA. 2005;102(19):6990–6995. doi: 10.1073/pnas.0500466102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Perez M, Hernandez F, Lim F, Diaz-Nido J, Avila J. Chronic lithium treatment decreases mutant tau protein aggregation in a transgenic mouse model. J Alzheimers Dis. 2003;5(4):301–308. doi: 10.3233/jad-2003-5405. [DOI] [PubMed] [Google Scholar]
- 125.Engel T, Goni-Oliver P, Lucas JJ, Avila J, Hernandez F. Chronic lithium administration to FTDP-17 tau and GSK-3β overexpressing mice prevents tau hyperphosphorylation and neurofibrillary tangle formation, but pre-formed neurofibrillary tangles do not revert. J Neurochem. 2006;99(6):1445–1455. doi: 10.1111/j.1471-4159.2006.04139.x. [DOI] [PubMed] [Google Scholar]
- 126.Selenica ML, Jensen HS, Larsen AK, Pedersen ML, Helboe L, Leist M, Lotharius J. Efficacy of small-molecule glycogen synthase kinase-3 inhibitors in the postnatal rat model of tau hyperphosphorylation. Br J Pharmacol. 2007;152(6):959–979. doi: 10.1038/sj.bjp.0707471. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Liu F, Iqbal K, Grundke-Iqbal I, Hart GW, Gong CX. O-GlcN Acylation regulates phosphorylation of tau: A mechanism involved in Alzheimer's disease. Proc Natl Acad Sci USA. 2004;101(29):10804–10809. doi: 10.1073/pnas.0400348101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Lefebvre T, Ferreira S, Dupont-Wallois L, Bussiere T, Dupire MJ, Delacourte A, Michalski JC, Caillet-Boudin ML. Evidence of a balance between phosphorylation and O-GlcNAc glycosylation of tau proteins - A role in nuclear localization. Biochim Biophys Acta. 2003;1619(2):167–176. doi: 10.1016/s0304-4165(02)00477-4. [DOI] [PubMed] [Google Scholar]
- 129.Bulic B, Pickhardt M, Schmidt B, Mandelkow EM, Waldmann H, Mandelkow E. Development of tau aggregation inhibitors for Alzheimer's disease. Angew Chem Int Ed Engl. 2009;48(10):1740–1752. doi: 10.1002/anie.200802621. [DOI] [PubMed] [Google Scholar]
- 130.Staff RT, Ahearn TS, Murray AD, Bentham P, Seng KM, Wischik C. P4-347: Tau aggregation inhibitor (TAI) therapy with Rember arrests the trajectory of rCBF decline in brain regions affected by tau pathology in mild and moderate Alzheimer's disease (AD) Alzheimers Dement. 2008;4:T775. [Google Scholar]
- 131.Pickhardt M, Gazova Z, von Bergen M, Khlistunova I, Wang Y, Hascher A, Mandelkow EM, Biernat J, Mandelkow E. Anthraquinones inhibit tau aggregation and dissolve Alzheimer's paired helical filaments in vitro and in cells. J Biol Chem. 2005;280(5):3628–3635. doi: 10.1074/jbc.M410984200. [DOI] [PubMed] [Google Scholar]
- 132.Larbig G, Pickhardt M, Lloyd DG, Schmidt B, Mandelkow E. Screening for inhibitors of tau protein aggregation into Alzheimer paired helical filaments: A ligand based approach results in successful scaffold hopping. Curr Alzheimer Res. 2007;4(3):315–323. doi: 10.2174/156720507781077250. [DOI] [PubMed] [Google Scholar]
- 133.Crowe A, Huang W, Ballatore C, Johnson RL, Hogan AM, Huang R, Wichterman J, McCoy J, Huryn D, Auld DS, Smith AB, 3rd, et al. Identification of aminothienopyridazine inhibitors of tau assembly by quantitative high-throughput screening. Biochemistry. 2009;48(32):7732–7745. doi: 10.1021/bi9006435. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Ravikumar B, Sarkar S, Berger Z, Rubinsztein DC. The roles of the ubiquitin-proteasome and autophagy-lysosome pathways in Huntington's disease and related conditions. Clin Neurosci Res. 2003;3(3):141–148. [Google Scholar]
- 135.Dickey CA, Kamal A, Lundgren K, Klosak N, Bailey RM, Dunmore J, Ash P, Shoraka S, Zlatkovic J, Eckman CB, Patterson C, et al. The high-affinity Hsp90-CHIP complex recognizes and selectively degrades phosphorylated tau client proteins. J Clin Invest. 2007;117(3):648–658. doi: 10.1172/JCI29715. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Zhang H, Burrows F. Targeting multiple signal transduction pathways through inhibition of Hsp90. J Mol Med. 2004;82(8):488–499. doi: 10.1007/s00109-004-0549-9. [DOI] [PubMed] [Google Scholar]
- 137.Dickey CA, Dunmore J, Lu B, Wang JW, Lee WC, Kamal A, Burrows F, Eckman C, Hutton M, Petrucelli L. Hsp induction mediates selective clearance of tau phosphorylated at proline-directed Ser/Thr sites but not KXGS (MARK) sites. FASEB J. 2006;20(6):753–755. doi: 10.1096/fj.05-5343fje. [DOI] [PubMed] [Google Scholar]
- 138.Luo W, Dou F, Rodina A, Chip S, Kim J, Zhao Q, Moulick K, Aguirre J, Wu N, Greengard P, Chiosis G. Roles of heat-shock protein 90 in maintaining and facilitating the neurodegenerative phenotype in tauopathies. Proc Natl Acad Sci USA. 2007;104(22):9511–9516. doi: 10.1073/pnas.0701055104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Berger Z, Ravikumar B, Menzies FM, Oroz LG, Underwood BR, Pangalos MN, Schmitt I, Wullner U, Evert BO, O'Kane CJ, Rubinsztein DC. Rapamycin alleviates toxicity of different aggregate-prone proteins. Hum Mol Genet. 2006;15(3):433–442. doi: 10.1093/hmg/ddi458. [DOI] [PubMed] [Google Scholar]
- 140.Hamano T, Gendron TF, Causevic E, Yen SH, Lin WL, Isidoro C, Deture M, Ko LW. Autophagic-lysosomal perturbation enhances tau aggregation in transfectants with induced wild-type tau expression. Eur J Neurosci. 2008;27(5):1119–1130. doi: 10.1111/j.1460-9568.2008.06084.x. [DOI] [PubMed] [Google Scholar]
- 141.Du H, Guo L, Fang F, Chen D, Sosunov AA, McKhann GM, Yan Y, Wang C, Zhang H, Molkentin JD, Gunn-Moore FJ, et al. Cyclophilin D deficiency attenuates mitochondrial and neuronal perturbation and ameliorates learning and memory in Alzheimer's disease. Nat Med. 2008;14(10):1097–1105. doi: 10.1038/nm.1868. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Halestrap AP, McStay GP, Clarke SJ. The permeability transition pore complex: Another view. Biochimie. 2002;84(2–3):153–166. doi: 10.1016/s0300-9084(02)01375-5. [DOI] [PubMed] [Google Scholar]
- 143.Crompton M. Mitochondria and aging: A role for the permeability transition? Aging Cell. 2004;3(1):3–6. doi: 10.1046/j.1474-9728.2003.00073.x. [DOI] [PubMed] [Google Scholar]
- 144.Halestrap A. Biochemistry: A pore way to die. Nature. 2005;434(7033):578–579. doi: 10.1038/434578a. [DOI] [PubMed] [Google Scholar]
- 145.Bachurin S, Bukatina E, Lermontova N, Tkachenko S, Afanasiev A, Grigoriev V, Grigorieva I, Ivanov Y, Sablin S, Zefirov N. Antihistamine agent Dimebon as a novel neuroprotector and a cognition enhancer. Ann N Y Acad Sci. 2001;939:425–435. doi: 10.1111/j.1749-6632.2001.tb03654.x. [DOI] [PubMed] [Google Scholar]
- 146.Lermontova NN, Redkozubov AE, Shevtsova EF, Serkova TP, Kireeva EG, Bachurin SO. Dimebon and tacrine inhibit neurotoxic action of β-amyloid in culture and block L-type Ca2+ channels. Bull Exp Biol Med. 2001;132(5):1079–1083. doi: 10.1023/a:1017972709652. [DOI] [PubMed] [Google Scholar]
- 147.Bachurin SO, Shevtsova EP, Kireeva EG, Oxenkrug GF, Sablin SO. Mitochondria as a target for neurotoxins and neuroprotective agents. Ann N Y Acad Sci. 2003;993:334–344. doi: 10.1111/j.1749-6632.2003.tb07541.x. [DOI] [PubMed] [Google Scholar]
- 148.Grigorev VV, Dranyi OA, Bachurin SO. Comparative study of action mechanisms of Dimebon and memantine on AMPA- and NMDA-subtypes glutamate receptors in rat cerebral neurons. Bull Exp Biol Med. 2003;136(5):474–477. doi: 10.1023/b:bebm.0000017097.75818.14. [DOI] [PubMed] [Google Scholar]
- 149. Doody RS, Gavrilova SI, Sano M, Thomas RG, Aisen PS, Bachurin SO, Seely L, Hung D. Effect of Dimebon on cognition, activities of daily living, behaviour, and global function in patients with mild-to-moderate Alzheimer's disease: A randomised, double-blind, placebo-controlled study. Lancet. 2008;372(9634):207–215. doi: 10.1016/S0140-6736(08)61074-0.. • The well-tolerated antihistamine latrepirdine is a promising treatment option for improving cognition in patients with AD. The mode of action of this agent may be through the protection of mitochondrial function.
- 150.Wu TY, Chen CP. Dual action of memantine in Alzheimer disease: A hypothesis. Taiwan J Obstet Gynecol. 2009;48(3):273–277. doi: 10.1016/S1028-4559(09)60303-X. [DOI] [PubMed] [Google Scholar]
- 151.Doraiswamy PM, Xiong GL. Pharmacological strategies for the prevention of Alzheimer's disease. Expert Opin Pharmacother. 2006;7(1):1–10. doi: 10.1517/14656566.7.1.1. [DOI] [PubMed] [Google Scholar]
- 152.Farlow MR. Utilizing combination therapy in the treatment of Alzheimer's disease. Expert Rev Neurother. 2004;4(5):799–808. doi: 10.1586/14737175.4.5.799. [DOI] [PubMed] [Google Scholar]
- 153.Jorm AF, Jolley D. The incidence of dementia: A meta-analysis. Neurology. 1998;51(3):728–733. doi: 10.1212/wnl.51.3.728. [DOI] [PubMed] [Google Scholar]
- 154.Manly JJ, Merchant CA, Jacobs DM, Small SA, Bell K, Ferin M, Mayeux R. Endogenous estrogen levels and Alzheimer's disease among postmenopausal women. Neurology. 2000;54(4):833–837. doi: 10.1212/wnl.54.4.833. [DOI] [PubMed] [Google Scholar]
- 155.Bonda DJ, Castellani RJ, Lee HG, Tabaton M, Casadesus G, Perry G, Zhu X, Smith MA. Down syndrome and Alzheimer disease: A 'multi-hit' hypothesis based on multiple contributing factors resulting in pleotrophic consequences. In: Jelinek D, Dvorak G, editors. Handbook of Down Syndrome Research. Hauppauge, NY, USA: Nova Science Publishers; 2010. in press. [Google Scholar]
- 156. Casadesus G, Webber KM, Atwood CS, Pappolla MA, Perry G, Bowen RL, Smith MA. Luteinizing hormone modulates cognition and amyloid-β deposition in Alzheimer APP transgenic mice. Biochim Biophys Acta. 2006;1762(4):447–452. doi: 10.1016/j.bbadis.2006.01.008.. • Hormone regulation is also being studied as a protective factor against the clinical and pathological effects of AD.
- 157.Short RA, Bowen RL, O'Brien PC, Graff-Radford NR. Elevated gonadotropin levels in patients with Alzheimer disease. Mayo Clin Proc. 2001;76(9):906–909. doi: 10.4065/76.9.906. [DOI] [PubMed] [Google Scholar]
- 158.Bowen RL, Isley JP, Atkinson RL. An association of elevated serum gonadotropin concentrations and Alzheimer disease? J Neuroendocrinol. 2000;12(4):351–354. doi: 10.1046/j.1365-2826.2000.00461.x. [DOI] [PubMed] [Google Scholar]
- 159.Bowen RL, Smith MA, Harris PL, Kubat Z, Martins RN, Castellani RJ, Perry G, Atwood CS. Elevated luteinizing hormone expression colocalizes with neurons vulnerable to Alzheimer's disease pathology. J Neurosci Res. 2002;70(3):514–518. doi: 10.1002/jnr.10452. [DOI] [PubMed] [Google Scholar]
- 160.Bowen RL, Verdile G, Liu T, Parlow AF, Perry G, Smith MA, Martins RN, Atwood CS. Luteinizing hormone, a reproductive regulator that modulates the processing of amyloid-β precursor protein and amyloid-β deposition. J Biol Chem. 2004;279(19):20539–20545. doi: 10.1074/jbc.M311993200. [DOI] [PubMed] [Google Scholar]
- 161.Sims NR, Bowen DM, Allen SJ, Smith CC, Neary D, Thomas DJ, Davison AN. Presynaptic cholinergic dysfunction in patients with dementia. J Neurochem. 1983;40(2):503–509. doi: 10.1111/j.1471-4159.1983.tb11311.x. [DOI] [PubMed] [Google Scholar]
- 162.DeKosky ST, Harbaugh RE, Schmitt FA, Bakay RA, Chui HC, Knopman DS, Reeder TM, Shetter AG, Senter HJ, Markesbery WR. Cortical biopsy in Alzheimer's disease: Diagnostic accuracy and neurochemical, neuropathological, and cognitive correlations. Intraventricular Bethanecol Study Group. Ann Neurol. 1992;32(5):625–632. doi: 10.1002/ana.410320505. [DOI] [PubMed] [Google Scholar]
- 163.Kasa P, Rakonczay Z, Gulya K. The cholinergic system in Alzheimer's disease. Prog Neurobiol. 1997;52(6):511–535. doi: 10.1016/s0301-0082(97)00028-2. [DOI] [PubMed] [Google Scholar]
- 164.Giacobini E. Cholinesterase inhibitors stabilize Alzheimer's disease. Ann N Y Acad Sci. 2000;920:321–327. doi: 10.1111/j.1749-6632.2000.tb06942.x. [DOI] [PubMed] [Google Scholar]
- 165.Giacobini E. Do cholinesterase inhibitors have disease-modifying effects in Alzheimer's disease? CNS Drugs. 2001;15(2):85–91. doi: 10.2165/00023210-200115020-00001. [DOI] [PubMed] [Google Scholar]
- 166.Giacobini E. Long-term stabilizing effect of cholinesterase inhibitors in the therapy of Alzheimer' disease. J Neural Transm Suppl. 2002;(62):181–187. doi: 10.1007/978-3-7091-6139-5_17. [DOI] [PubMed] [Google Scholar]