Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2023 Apr 29.
Published in final edited form as: CNS Drugs. 2016 Aug;30(8):667–675. doi: 10.1007/s40263-016-0364-1

Aβ-degrading proteases:therapeutic potential in Alzheimer disease

Malcolm A Leissring 1,*
PMCID: PMC10148599  NIHMSID: NIHMS1892567  PMID: 27349988

Abstract

The amyloid ß-protein (Aß) is well established to play an indispensible role in the pathogenesis of Alzheimer disease (AD). Aβ is subject to proteolytic degradation by a diverse array of peptidases and proteinases, known collectively as Aβ-degrading proteases (AβDPs). A growing number of AβDPs have been identified, which impact Aß powerfully and in a surprising variety of ways. As such, AßDPs hold considerable therapeutic potential for the treatment and/or prevention of AD. Here we critically review the relative merits of therapeutic strategies targeting AßDPs as compared to current Aß-lowering strategies focused on immunotherapies and pharmacological modulation of Aß-producing enzymes. Several innovative advances have increased considerably the feasibility of delivering AßDPs to the brain or enhancing their activity in a non-invasive manner. We argue that therapies targeting AßDPs offer numerous potential advantages, which should be explored through continued research into this promising field.

Introduction

Alzheimer disease (AD) is a debilitating and increasingly common neurodegenerative disorder that results in progressive loss of memory, cognition, language skills, and personality traits [1]. Histopathologically, the disease is characterized by progressive accumulation of “plaques” composed of the amyloid-ß protein (Aß) and “tangles” comprised of hyperphosphorylated forms of the microtubule-associated protein, tau [1]. Currently, over 35 million people worldwide are afflicted [2], including 5.3 million individuals in the USA [3]. As people are living longer and the leading edge of the baby-boomer generation ages, AD is growing to epidemic proportions. Recent studies indicate that AD affects a larger portion of the population than previously believed, now ranking as the 3rd leading cause of death, exceeded only by heart disease and cancer [4]. The financial burden imposed by the marked increase in AD cases, as well as the stress on caregivers and family members, represents an unprecedented global public health challenge. Alzheimer’s Disease International (ADI) estimates a global annual cost of dementia, of which AD is the leading cause, of ~$818 billion or 1% of global GDP [2]. To date, over 100 AD clinical trials have been conducted [5] and—sadly—none has proven effective at halting or even slowing disease progression significantly. There is, therefore, a vital need to investigate alternative treatment strategies that can prevent or treat this debilitating disease.

Centrality of Aß mismetabolism

Advances in human molecular genetics have contributed inestimably to our understanding of the molecular pathogenesis of AD, demonstrating that specific perturbations to Aß metabolism are sufficient to trigger the full spectrum of the disease [6]. However, the cause of AD is known with certainty in only a tiny minority (~1%) of all patients, specifically early-onset, familial AD (FAD) cases attributable to dominant-negative mutations in 3 genes: the amyloid precursor protein (APP) and presenilin 1 and 2 (APP, PSEN1, PSEN2) [7]. Several hundred FAD-linked mutations have been identified, and all universally increase either total Aß levels or specifically the relative abundance of longer, more pathogenic forms of Aß such as Aß42 [8]. Individuals with Down syndrome (trisomy 21), who carry 3 copies of the APP gene on chromosome 21 and therefore produce 50% more Aß than normal, also invariably develop AD-type pathology [8]. Collectively, these genetic findings provide exceptionally strong evidence that perturbed Aß metabolism plays a causal role in triggering the full spectrum of AD-type pathology, including those elements (e.g., NFTs and neurodegeneration) that only appear at later stages in the disease [7].

For the vast majority of AD cases, however—commonly referred to as sporadic, idiopathic or late-onset AD—Aß accumulates abnormally, but the operative mechanisms remain less certain. Increases in Aß production not attributable to FAD-linked mutations have been documented to occur in some AD patients [9, 10]). However, it has been hypothesized that reduced clearance of Aß—either alternatively or in addition to increased Aß production—may be operative in the etiology of many cases of sporadic AD [6, 11]. Consistent with this idea, allelic variations in apolipoprotein E (ApoE), which is implicated in the clearance of Aß, dramatically modulate AD risk [12].

The hypothesis that impaired Aß clearance per se is occurring in some sporadic AD cases remained largely speculative for many decades. In recent years, however, powerful techniques have been developed that, through the transient administration of radiolabeled amino acids to human subjects, permit the accurate quantification of the rates of Aß production and clearance in the cerebrospinal fluid (CSF) of living patients [13]. These elegant studies have confirmed that Aß clearance—or more accurately, the rate of Aß clearance relative to that of production—is indeed decreased in sporadic AD cases [14].

Lessons from therapies targeting Aß production

Given the certitude that excessive production of Aß causes familial forms of AD, it is unsurprising that most Aß-lowering therapeutics strategies under development target the enzymes that mediate Aß production (or, as considered in a subsequent section, Aß itself). Aß is generated from APP, a type 1 membrane protein, via the action of proteolytic enzymes known as “secretases” because their action results in the secretion of Aß and/or the large extracellular ectodomain of APP [15]. In the processing pathway that results in Aß production, APP is initially cleaved by ß-secretase, an aspartic protease known as beta-site APP-cleaving enzyme 1 (BACE1) [15], releasing a fragment known as sAPPß. The remaining membrane-bound C-terminal fragment (termed C99) is subsequently cleaved by γ-secretase, an intramembraneous complex of four proteins, the active-site of which is formed by presenilin-1 or −2 [15].

Two aspects of γ-secretase are noteworthy: first, it cuts at positions within the membrane, an unusual site for hydrolysis to take place. Second, unlike most proteases, γ-secretase does not cut a specific peptide bond, but instead cleaves at any of several sites, resulting in the generation of a mixture of Aß species from 37 to 48 amino acids in length, with notable examples being Aß40 and Aß42 [15]. FAD-linked mutations in presenilins cause the production of Aß42 relative to Aß40 (the Aß42/40 ratio) to increase [16], and this shift that has been strongly implicated in the pathogenesis of FAD [17, 18].

Once the mechanistic details of Aß processing were elucidated, it became clear that there were two fundamental ways to reduce Aß production: via inhibition of BACE1 or the presenilin/γ-secretase complex. Outright inhibition of these proteolytic activities, however, has proved to be problematic, primarily because they process numerous substrates besides APP. For example, although genetic deletion of BACE1 in mice results in viable animals that exhibit absolute cessation of Aß production [19, 20], several more subtle, but worrisome phenotypes, such as hypomyelination, were revealed upon closer analysis [21]. More recently, ß-secretase has been shown to be an active sheddase in mouse primary neurons, where it contributes to the secretion of approximately one fifth of identified shed proteins [22], including neuregulin, which has important functions in myelination [23]. Furthermore, chronic pharmacological inhibition of BACE1 has resulted in adverse effects in mice [24]. Finally, potential secondary effects have been observed in clinical trials of BACE1 inhibitors [25].

Pharmacologic inhibition of presenilin/γ-secretase has proved to be problematic, as well, for similar reasons. In mice, genetic deletion of presenilin-1 results in premature lethality [26], attributable in part to impairments the processing of notch1, an intramembrane protein involved in cell fate determination during development [27]. As was true for BACE1, numerous substrates for presenilin/γ-secretase besides APP and notch1 have been identified over the years [28], raising further questions about the viability of outright inhibition of γ-secretase. Given this, it is perhaps unsurprising that the γ-secretase inhibitor (GSI) semagacestat [29], although effective at lowering Aß in humans [30], actually resulted in cognitive worsening when tested in long-term clinical trials [31, 29].

Notwithstanding the theoretical and empirical problems with GSIs, there is considerable hope for a class of compounds known as γ-secretase modulators (GSMs). Due to the complexity and unusual structure of the presenilin/γ-secretase complex [32], compounds have been identified that modulate, rather than block, γ-secretase activity in both substrate-specific and cleavage site-specific ways [33]. Thus, it has been possible to develop “notch-sparing” inhibitors of APP processing [33], as well as compounds that specifically reduce the Aß42/40 ratio [34]. Although GSMs hold perhaps the greatest promise among those therapies targeting Aß production, there is some concern about focusing too narrowly on Aß- and notch-specific effects, to the exclusion of possible influences on the many other substrates processed by presenilin/γ-secretase. This caveat is especially relevant given accumulating evidence that the pathogenicity of certain FAD presenilin mutations may result from an overall decrease in γ-secretase function in addition to their effect on the Aß42/40 ratio [35].

Lessons from Aß immunotherapies

The other approach to lowering Aß that has been extensively explored is that of enhancing clearance of cerebral Aß through active or passive immunization with antibodies against Aß. Like therapies targeting AßDPs, discussed below, this approach has the advantage of circumventing the deleterious effects associated with ß- and γ-secretase inhibition. Active immunization proved highly effective in removing brain Aß in mouse models [3640] as well as humans [41], and early studies showed some evidence of blunting cognitive decline in some AD patients [42]. However, this approach also triggered deleterious immune-mediated reactions in a subset of patients [43]. To overcome these problems, considerable effort has explored the therapeutic potential of passive immunization, wherein the patient is administered humanized monoclonal antibodies against Aß. Unlike active immunization, this approach has the advantage of being reversible and also avoiding the deleterious consequences of cell-mediated immunity [44]. Nevertheless, despite a number of ambitious clinical trials, none have proven unambiguously to blunt cognitive decline. Moreover, concerns have been raised about the ability of Aß antibodies to translocate Aß from the parenchyma to the brain vasculature, together with concerns about pro-inflammatory responses triggered by the Fc portion of anti-Aß antibodies [45]. Thus, despite the seemingly great promise of Aß immunotherapy, sufficient uncertainty about the efficacy and safety of this approach remains to warrant continued exploration of alternative therapeutic strategies.

General features of AßDPs

Given the lack of success obtained with Aß immunotherapies and drugs targeting Aß production, it is reasonable to consider whether therapies targeting AßDPs might prove to be viable—or even superior—as alternatives for lowering of brain Aß. As a general class, it is difficult to overstate the importance of AßDPs for the regulation of cerebral Aß levels. It has been estimated that over 8 decades of life the average individual generates >10 kg of Aß [11]; since only a tiny fraction of Aß incorporates into insoluble aggregates such as plaques or cerebral amyloid, it follows that the overwhelming majority of Aß peptides produced are ultimately destroyed by AßDPs. In a particularly striking illustration, inhibition of just a subset of just one class of proteases (zinc-metalloproteases) via i.c.v. administration of phosphoramidon produced a >400% increase in cerebral Aß within 30 min [46, 47]. When compared, for example, to trisomy 21, wherein a mere 50% increase in Aß gives rise to AD-type pathology by the 4th decade of life, it is easy to see how even a partial impairment in Aß degradation could play a significant role in AD pathogenesis.

It is important to emphasize that proteolytic degradation of Aß represents just one of several mechanisms of Aß clearance that operate in parallel. Cerebral Aß levels are also regulated by such processes as cell-mediated clearance and active and passive transport of Aß across the blood-brain barrier [48]. It is difficult to rank the relative importance of these different clearance mechanisms, in no small part because they ultimately culminate in proteolytic degradation (e.g., via lysosomal proteases). From a therapeutic perspective, however, these highly complex pathways may be more difficult to manipulate than AßDPs, which represent discrete therapeutic targets.

AßDPs can be sorted into several broad functional categories (Table 1). Endogenous regulators of Aß are AßDPs involved in the regulation of Aß under physiological conditions. Examples include insulin-degrading enzyme (IDE), neprilysin, and endothelin-converting enzymes-1 and −2 (ECE1; ECE2) [49]. By definition, deletion or inhibition of an endogenous regulator will necessarily produce elevations in cerebral Aß in the absence of pathology. It is notable that a large number of AßDPs fit this category, because this demonstrates that Aß clearance is operating at capacity, such that the elimination of any of several AßDPs will increase cerebral Aß. By the same token, increasing the activity or levels of any AßDP—including one not normally involved in Aß catabolism—will have the effect of lowering Aß levels; a finding that has been demonstrated multiple times in mouse models.

Table 1.

Properties of known Aß-degrading proteases

Type Protease Substratesa Functional Categoryb Subcellular localizationc
Oligos Fibrils
Metallo NEP synth no E,P,T ex, ER, G
NEP2 E ex, ER, G
hMMEL ex, ER, G
ECE1 E ex, ER, G, endo
ECE2 E ex, ER, G, endo
ACE ex, ER, G
MMP2 yes ex, ER, G
MMP9 yes ex, ER, G
CD147/EMMPRIN ex, ER, G, endo
IDE no no E,T ex, ER, endo, lyso, mito
PreP mito
Serine Plasmin/tPA/uPA natural yes P,T ex, ER, G
Acylpeptide hydrolase cyto
Myelin basic protein ex, ER, G
Aspartate Cathepsin D yes E endo, lyso
BACE1 T endo, lyso
BACE2 No E endo, lyso
Cysteine Cathepsin B E,T ex, endo, lyso
Threonine Proteasome cyto
a

Where known, indicates whether the corresponding AßDP degrades oligomeric (ologos) or fibrillar (fibrils) Aß.

b

Where known, indicates functional category of each AßDP (E, endogenous regulator; P, pathologic regulator; T, therapeutic regulator).

c

Indicates subcellular localization (cyto, cytosol; endo, endosomes; ER, endoplasmic reticulum; ex, extracellular space; G, Golgi network; lyso, lysosomes; mito, mitochondria). ACE, angiotensin-converting enzyme; BACE1,2, beta-site APP-cleaving enzyme 1, 2; ECE1,2, endothelin-converting enzyme 1,2; EMMPRIN, extracellular matrix metalloproteinase inducer; hMMEL, human membrane metallo-endopeptidase-like protein; IDE, insulin-degrading enzyme; MMP2,9, matrix-metalloproteinase 2,9; NEP, neprilysin; NEP2 neprilysin 2; PreP, presequence peptidase; tPA, tissue-type plasminogen activator; uPA, urokinase-type plasminogen activator. For primary references, see ref. [50].

The second functional category is pathological regulators, which are AßDPs that are only active in the presence of overt Aß pathology. Plasmin is prime example of a pathological regulator [49]: in the absence of pathology, deletion of the precursor protein for plasmin (plasminogen) gives rise to no net increase in Aß; however, in the presence of aggregated forms of Aß, plasminogen get converted to active plasmin by tissue-type plasminogen activator (tPA), a protease that is sensitive to the presence of beta-pleated sheets.

A final category might be termed therapeutic regulators. Although any of the above proteases could be used therapeutically, it is critical to recognize that any protease capable of degrading Aß—even an engineered protease or one not normally expressed in brain—could in principle be used therapeutically [50]. Thus, as compared to the mere two proteases involved in Aß production, it is apparent that AßDPs represent and enormously diverse collection of possible therapeutic targets, which have only barely been explored for their therapeutic potential.

AßDPs can be further distinguished in terms of their proteolytic properties and the substrates they can act on. As is true for all proteases, AßDPs can include endoproteinases, aminopeptidases and carboxypeptidases. The latter class is of special interest, since certain proteases—such as cathepsin B—can convert the longer, more pathogenic forms of Aß, such as Aß42, to shorter species, such as Aß40 or Aß38, via carboxypeptidase activity [51]. Finally, given that Aß can exist in monomeric forms as well as multiple distinct aggregated forms [52, 53], ranging from dimers and trimers, to a hypothesized dodecameric species termed Aß*56, to protofibrils, fibrils, and, finally, as macroscopic “plaques,” AßDPs can be categorized in terms of which Aß species they are capable of hydrolyzing (Table 1). For example, pure peptidases, such as IDE, can act only on monomeric Aß species, while other AßDPs can degrade oligomeric or fibrillar forms of Aß [50].

The question of which species/conformations of Aß different AßDPs target may be of particular therapeutic relevance. For example, high-level transgenic overexpression of neprilysin in a mouse model of AD has been shown to prevent the formation of extracellular amyloid deposits completely [54]. Nevertheless, oligomeric Aß—a particularly neurotoxic form of Aß [52]—was found to be unchanged by neprilysin overexpression in these same animals; moreover, the mice lacking plaques (but containing oligomers) also showed cognitive defects identical to APP transgenic mice not overexpressing neprilysin [55]. In a related vein, by virtue of their particular subcellular localization profiles and pH optima, different AßDPs can target spatially distinct “pools” of Aß (e.g., extracellular, endosomal, lysosomal) (Table 1) [56]. As is true for the various species of Aß, some pools of Aß are more strongly implicated than others in AD pathogenesis [57, 58]. Intriguingly, there is some evidence that extracellular pools of Aß may not be the most relevant to cognitive defects. For instance, overexpression of Aß42 exclusively in the extracellular space (using BRI-Aß42 fusion proteins [59]) led to the development of robust amyloid plaques [60], but failed to affect cognitive performance [61]. These intriguing findings could explain why therapies that target extracellular Aß, such as immunotherapies—have failed to impact cognitive decline.

Several additional intrinsic features of AßDPs render them particularly attractive as therapeutic targets. First and foremost, the general strategy of targeting the removal of Aß after it is produced obviates all of the aforementioned concerns about the machinery involved in Aß production. Moreover, relative to other approaches, such as immunotherapies or inhibitors of Aß aggregation, which necessarily act stoichiometrically, AßDPs have the distinct advantage of acting catalytically and irreversibly to remove Aß [62]. Thus, a very large reduction in Aß levels can be effected by a relatively small increase in the level or activity of a specific AßDP [62]. Finally, whereas Aß production necessarily occurs exclusively in the proximity of ß- and γ-secretase, Aß degradation can take place any arbitrary distance from the sites of Aß production [56]. Indeed, some reports have suggested that increasing Aß degradation (or sequestration [63]) in the periphery can lower cerebral Aß levels [64, 65], although these initial results were not confirmed in several well designed studies [66, 67]. Nevertheless, from a drug development perspective, the fact that AßDP-targeting therapies can operate at some distance from the source of Aß production represents a distinct advantage over existing Aß-lowering strategies.

Because AßDPs degrade a number of substrates besides Aß, it is important to consider the potential off-target effects of increasing particular AßDPs. Although this is a valid concern, it should be noted that AßDPs are in no way unique in this regard; the secretases involved in Aß production also degrade numerous substrates besides APP, yet this has not prevented secretase inhibitors from being intensively investigated in clinical trials. Moreover, as is true for γ-secretase, there is strong evidence that certain AßDPs can be modulated by small molecules in a highly substrate-specific manner [68, 69]. Furthermore, if certain forms of AD are attributable to deficiencies in specific AßDPs, restoring the activity of that AßDP to normal levels would not be expected to have side effects. Finally, animal modeling studies involving high-levels overexpression of NEP or IDE have not revealed overtly deleterious consequences [54, 70, 71, 55]. Nevertheless, the potential for off-target effects is an important one, and it will therefore be of critical importance to comprehensively evaluate the potential side effects of candidate therapeutic AßDPs in animal studies prior to any clinical applications.

Therapeutic approaches to targeting AßDPs

At first glance, AßDPs would appear to be most amenable to gene therapeutic approaches. Animal modeling studies have confirmed the viability of this approach. Long-term viral-mediated expression of neprilysin, for example, has been shown to lower Aß levels and ameliorate Aß-associated cognitive deficits [71]. However, there remain serious theoretical and practical concerns about the clinical use of potentially risky viral vectors and associated invasive procedures to deliver them. Nevertheless, gene therapies are already being tested in AD patients. For instance, a clinical trial of nerve growth factor gene delivery was conducted in 2001 [72], and post-mortem analysis revealed significant effects lasting as long as 10 years [73]. It is notable, however, that two of the patients in this trial were severely affected by the intrusive surgical procedure, one of whom died [72]. Numerous advances have been made in the intervening years, however, that suggest less-invasive means of gene-delivery might be feasible [74]. For example, viral delivery of neprilysin in rats has been shown to be enhanced significantly using convention-enhanced delivery [75]. A modified adeno-associated virus (AAV) vector has been developed by Saido and colleagues that produced neuronal expression of neprilysin throughout the brain of mice following peripheral delivery [76].

Delivery of AßDPs to the CNS does not necessarily require the administration of viral vectors or genetically modified cells. For example, Masliah and colleagues developed a recombinant form of neprilysin [76] containing a brain-transport peptide [77]. In initial studies, this fusion protein was delivered via a lentiviral vector administered peripherally and expressed primarily in liver and spleen [76]. Using this approach, the recombinant neprilysin was shown to effectively lower cerebral Aß levels, plaque burden and also reverse cognitive deficits. Notably, in subsequent studies this team succeeded in obtaining similarly positive results via peripheral administration of the recombinant fusion protein alone [78]. Thus, with sufficient improvements, it may be feasible to deliver AßDPs intravenously, in similar manner to Aß immunotherapies.

Can small molecules be developed that increase the activity of AßDPs? Generally speaking, it is widely assumed that it is not feasible to increase the activity of a protease, but this is not, in fact, true. For example, many proteases are regulated by endogenous inhibitors, and it is possible to develop compounds that block the interaction of the protease and its inhibitor, thereby disinhibiting the protease. For example, Wyeth developed a small-molecule activator of plasmin which showed efficacy in an animal model of AD [79]. This compound acted by displacing plasminogen activator inhibitor-1 (PAI1), an endogenous inhibitor of tissue-type plasminogen activator (tPA), which converts plasminogen to plasmin [79]. Since, as mentioned above, tPA is activated by beta-pleated sheets, including those formed by Aß [80], this compound effectively lowered the threshold for tPA-mediated plasmin activation by Aß. Direct activators of other enzymes [81], including AßDPs such as IDE [68, 82], have also been identified. Thus, the “druggability” of AßDPs is greater than has been widely assumed.

Rather than pharmacologically activating AßDPs, it is also possible to increase their expression. For example, levels of just a single AßDP, neprilysin, have been shown to increase in response to a wide variety of factors, including substance P [83], Aß [84, 85], to the intracellular domain of APP [86, 87], estrogen [88], and even environmental enrichment [89]. Saido and colleagues also found that administration of somatostatin reduced brain Aß levels via activation of neprilysin, and suggested that drugs targeting somatostatin receptors might be candidates as Aß-lowering targets [90]. Finally, given the great advantages of natural products in terms of safety and availability, it is notable that green tea extract has been shown to increase neprilysin levels [91]. Other dietary supplements have been shown to reduce Aß in animals models [92] and should be investigated for possible effects on AßDPs.

As is true for all Aß-lowering therapeutics, the question of when to initiate therapies targeting AßDPs remains critical. Basic research has not yet addressed this question directly, but the lack of success of clinical trials with other Aß-lowering approaches initiated in patients already showing memory loss suggests that it may be critical to intervene as early as possible in the course of the disease. Given the long-term nature of AD pathogenesis, manipulations that are comparably long-lasting, such as gene-therapeutic approaches, may therefore be particularly advantageous relative to most other, short-acting Aß-lowering therapeutics.

Conclusions

As a general class, AßDPs represent attractive targets for drug development. By virtue of their catalytic nature, AßDPs represent powerful regulators of Aß levels, and their sheer number and variety increases the chances that at least one will be found that can be targeted pharmacologically or expressed or administered therapeutically. Relative to the molecular machinery involved in Aß production, which has been the focus intensive research in both academia and industry, significantly less is known about AßDPs generally, including the complete set of AßDPs, and there have been no efforts to evaluate the risk-reward profiles of different AßDPs in a systematic fashion. Given the many novel aspects of AßDPs considered here, continued research into the possible therapeutic utility of AßDPs is clearly warranted.

Bullet point summary.

  • Proteases that degrade the amyloid ß-protein (Aß), which is central to the pathogenesis Alzheimer disease (AD), are powerful regulators of cerebral Aß levels and downstream cytopathological and cognitive sequelae.

  • By virtue of their sheer number and diversity, Aß-degrading proteases (AßDPs) offer a rich source of therapeutic targets to combat AD, with several intrinsic advantages over existing strategies to reducing cerebral Aß.

  • Recent advances have shown that the activity and/or levels of AßDPs can be enhanced through a variety of non-invasive therapeutic approaches.

Funding:

Supported by Grant No. 7–11-CD-06 from the American Diabetes Association to M.A.L. No funds were received specifically for the publication of this review.

Footnotes

Compliance with Ethical Standards

Conflicts of Interest: The author declares no competing interests.

References

  • 1.Mucke L. Neuroscience: Alzheimer’s disease. Nature. 2009;461(7266):895–7. [DOI] [PubMed] [Google Scholar]
  • 2.Prince M, Wimo A, Guerchet M, Ali GC, Wu TT, Prina M et al. World Alzheimer’s Report 2015. The global impact of dementia: an analysis of prevalence, incedence, cost and trends. London: Alzheimer’s Disease International; 2015. [Google Scholar]
  • 3.Alzheimer’s A. 2015 Alzheimer’s disease facts and figures. Alzheimer’s & dementia : the journal of the Alzheimer’s Association. 2015;11(3):332–84. [DOI] [PubMed] [Google Scholar]
  • 4.James BD, Leurgans SE, Hebert LE, Scherr PA, Yaffe K, Bennett DA. Contribution of Alzheimer disease to mortality in the United States. Neurology. 2014;82(12):1045–50. doi: 10.1212/WNL.0000000000000240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Godyn J, Jonczyk J, Panek D, Malawska B. Therapeutic strategies for Alzheimer’s disease in clinical trials. Pharmacological reports : PR. 2016;68(1):127–38. doi: 10.1016/j.pharep.2015.07.006. [DOI] [PubMed] [Google Scholar]
  • 6.Hardy J, Selkoe DJ. The amyloid hypothesis of Alzheimer’s disease: progress and problems on the road to therapeutics. Science. 2002;297(5580):353–6. doi: 10.1126/science.1072994. [DOI] [PubMed] [Google Scholar]
  • 7.Selkoe DJ, Schenk D. Alzheimer’s disease: molecular understanding predicts amyloid-based therapeutics. Annu Rev Pharmacol Toxicol. 2003;43:545–84. [DOI] [PubMed] [Google Scholar]
  • 8.Tanzi RE. The genetics of Alzheimer disease. Cold Spring Harbor perspectives in medicine. 2012;2(10). doi: 10.1101/cshperspect.a006296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Holsinger RM, McLean CA, Beyreuther K, Masters CL, Evin G. Increased expression of the amyloid precursor beta-secretase in Alzheimer’s disease. Ann Neurol. 2002;51(6):783–6. doi: 10.1002/ana.10208. [DOI] [PubMed] [Google Scholar]
  • 10.Holsinger RM, Lee JS, Boyd A, Masters CL, Collins SJ. CSF BACE1 activity is increased in CJD and Alzheimer disease versus [corrected] other dementias. Neurology. 2006;67(4):710–2. doi: 10.1212/01.wnl.0000229925.52203.4c. [DOI] [PubMed] [Google Scholar]
  • 11.Leissring MA. Proteolytic degradation of the amyloid beta-protein: the forgotten side of Alzheimer’s disease. Curr Alzheimer Res. 2006;3(5):431–5. [DOI] [PubMed] [Google Scholar]
  • 12.Bu G. Apolipoprotein E and its receptors in Alzheimer’s disease: pathways, pathogenesis and therapy. Nat Rev Neurosci. 2009;10(5):333–44. doi: 10.1038/nrn2620. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Mawuenyega KG, Kasten T, Sigurdson W, Bateman RJ. Amyloid-beta isoform metabolism quantitation by stable isotope-labeled kinetics. Anal Biochem. 2013;440(1):56–62. doi: 10.1016/j.ab.2013.04.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Mawuenyega KG, Sigurdson W, Ovod V, Munsell L, Kasten T, Morris JC et al. Decreased clearance of CNS beta-amyloid in Alzheimer’s disease. Science. 2010;330(6012):1774. doi:science.1197623 [pii] 10.1126/science.1197623. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.De Strooper B, Vassar R, Golde T. The secretases: enzymes with therapeutic potential in Alzheimer disease. Nat Rev Neurol. 2010;6(2):99–107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Selkoe DJ, Wolfe MS. Presenilin: running with scissors in the membrane. Cell. 2007;131(2):215–21. [DOI] [PubMed] [Google Scholar]
  • 17.Jankowsky JL, Fadale DJ, Anderson J, Xu GM, Gonzales V, Jenkins NA et al. Mutant presenilins specifically elevate the levels of the 42 residue beta-amyloid peptide in vivo: evidence for augmentation of a 42-specific gamma secretase. Hum Mol Genet. 2004;13(2):159–70. doi: 10.1093/hmg/ddh019. [DOI] [PubMed] [Google Scholar]
  • 18.Suzuki N, Cheung TT, Cai XD, Odaka A, Otvos L Jr., Eckman C et al. An increased percentage of long amyloid beta protein secreted by familial amyloid beta protein precursor (beta APP717) mutants. Science. 1994;264(5163):1336–40. [DOI] [PubMed] [Google Scholar]
  • 19.Luo Y, Bolon B, Damore MA, Fitzpatrick D, Liu H, Zhang J et al. BACE1 (beta-secretase) knockout mice do not acquire compensatory gene expression changes or develop neural lesions over time. Neurobiol Dis. 2003;14(1):81–8. doi:S0969996103001049 [pii]. [DOI] [PubMed] [Google Scholar]
  • 20.Luo Y, Bolon B, Kahn S, Bennett BD, Babu-Khan S, Denis P et al. Mice deficient in BACE1, the Alzheimer’s beta-secretase, have normal phenotype and abolished beta-amyloid generation. Nat Neurosci. 2001;4(3):231–2. doi: 10.1038/85059. [DOI] [PubMed] [Google Scholar]
  • 21.Cole SL, Vassar R. BACE1 structure and function in health and Alzheimer’s disease. Curr Alzheimer Res. 2008;5(2):100–20. [DOI] [PubMed] [Google Scholar]
  • 22.Kuhn PH, Koroniak K, Hogl S, Colombo A, Zeitschel U, Willem M et al. Secretome protein enrichment identifies physiological BACE1 protease substrates in neurons. EMBO J. 2012;31(14):3157–68. doi: 10.1038/emboj.2012.173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Fleck D, van Bebber F, Colombo A, Galante C, Schwenk BM, Rabe L et al. Dual cleavage of neuregulin 1 type III by BACE1 and ADAM17 liberates its EGF-like domain and allows paracrine signaling. J Neurosci. 2013;33(18):7856–69. doi: 10.1523/JNEUROSCI.3372-12.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Filser S, Ovsepian SV, Masana M, Blazquez-Llorca L, Brandt Elvang A, Volbracht C et al. Pharmacological inhibition of BACE1 impairs synaptic plasticity and cognitive functions. Biological psychiatry. 2015;77(8):729–39. doi: 10.1016/j.biopsych.2014.10.013. [DOI] [PubMed] [Google Scholar]
  • 25.Yan R, Vassar R. Targeting the beta secretase BACE1 for Alzheimer’s disease therapy. Lancet Neurol. 2014;13(3):319–29. doi: 10.1016/S1474-4422(13)70276-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Shen J, Bronson RT, Chen DF, Xia W, Selkoe DJ, Tonegawa S. Skeletal and CNS defects in Presenilin-1-deficient mice. Cell. 1997;89(4):629–39. [DOI] [PubMed] [Google Scholar]
  • 27.Selkoe DJ. Presenilin, Notch, and the genesis and treatment of Alzheimer’s disease. Proc Natl Acad Sci U S A. 2001;98(20):11039–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Lleo A, Saura CA. gamma-secretase substrates and their implications for drug development in Alzheimer’s disease. Current topics in medicinal chemistry. 2011;11(12):1513–27. [DOI] [PubMed] [Google Scholar]
  • 29.De Strooper B. Lessons from a failed gamma-secretase Alzheimer trial. Cell. 2014;159(4):721–6. doi: 10.1016/j.cell.2014.10.016. [DOI] [PubMed] [Google Scholar]
  • 30.Siemers ER, Dean RA, Friedrich S, Ferguson-Sells L, Gonzales C, Farlow MR et al. Safety, tolerability, and effects on plasma and cerebrospinal fluid amyloid-beta after inhibition of gamma-secretase. Clinical neuropharmacology. 2007;30(6):317–25. doi: 10.1097/WNF.0b013e31805b7660. [DOI] [PubMed] [Google Scholar]
  • 31.Ascher-Svanum H, Chen YF, Hake A, Kahle-Wrobleski K, Schuster D, Kendall D et al. Cognitive and Functional Decline in Patients With Mild Alzheimer Dementia With or Without Comorbid Diabetes. Clinical therapeutics. 2015;37(6):1195–205. doi: 10.1016/j.clinthera.2015.01.002. [DOI] [PubMed] [Google Scholar]
  • 32.Bai XC, Yan C, Yang G, Lu P, Ma D, Sun L et al. An atomic structure of human gamma-secretase. Nature. 2015;525(7568):212–7. doi: 10.1038/nature14892. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Wolfe MS. Inhibition and modulation of gamma-secretase for Alzheimer’s disease. Neurotherapeutics. 2008;5(3):391–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Eriksen JL, Sagi SA, Smith TE, Weggen S, Das P, McLendon DC et al. NSAIDs and enantiomers of flurbiprofen target gamma-secretase and lower Abeta 42 in vivo. J Clin Invest. 2003;112(3):440–9. doi: 10.1172/JCI18162. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.De Strooper B, Iwatsubo T, Wolfe MS. Presenilins and gamma-secretase: structure, function, and role in Alzheimer Disease. Cold Spring Harbor perspectives in medicine. 2012;2(1):a006304. doi: 10.1101/cshperspect.a006304. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Bard F, Cannon C, Barbour R, Burke RL, Games D, Grajeda H et al. Peripherally administered antibodies against amyloid beta-peptide enter the central nervous system and reduce pathology in a mouse model of Alzheimer disease. Nat Med. 2000;6(8):916–9. [DOI] [PubMed] [Google Scholar]
  • 37.DeMattos RB, Bales KR, Cummins DJ, Dodart JC, Paul SM, Holtzman DM. 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 U S A. 2001;98(15):8850–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Hock C, Konietzko U, Papassotiropoulos A, Wollmer A, Streffer J, von Rotz RC et al. Generation of antibodies specific for beta-amyloid by vaccination of patients with Alzheimer disease. Nat Med. 2002;8(11):1270–5. [DOI] [PubMed] [Google Scholar]
  • 39.Schenk D, Barbour R, Dunn W, Gordon G, Grajeda H, Guido T et al. Immunization with amyloid-beta attenuates Alzheimer-disease-like pathology in the PDAPP mouse. Nature. 1999;400(6740):173–7. [DOI] [PubMed] [Google Scholar]
  • 40.Weiner HL, Lemere CA, Maron R, Spooner ET, Grenfell TJ, Mori C et al. Nasal administration of amyloid-beta peptide decreases cerebral amyloid burden in a mouse model of Alzheimer’s disease. Ann Neurol. 2000;48(4):567–79. [PubMed] [Google Scholar]
  • 41.Holmes C, Boche D, Wilkinson D, Yadegarfar G, Hopkins V, Bayer A 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(9634):216–23. doi: 10.1016/S0140-6736(08)61075-2. [DOI] [PubMed] [Google Scholar]
  • 42.Hock C, Konietzko U, Streffer JR, Tracy J, Signorell A, Muller-Tillmanns B et al. Antibodies against beta-amyloid slow cognitive decline in Alzheimer’s disease. Neuron. 2003;38(4):547–54. [DOI] [PubMed] [Google Scholar]
  • 43.Nicoll JA, Wilkinson D, Holmes C, Steart P, Markham H, Weller RO. Neuropathology of human Alzheimer disease after immunization with amyloid-beta peptide: a case report. Nat Med. 2003;9(4):448–52. [DOI] [PubMed] [Google Scholar]
  • 44.Liu YH, Giunta B, Zhou HD, Tan J, Wang YJ. Immunotherapy for Alzheimer disease: the challenge of adverse effects. Nat Rev Neurol. 2012;8(8):465–9. doi: 10.1038/nrneurol.2012.118. [DOI] [PubMed] [Google Scholar]
  • 45.Fuller JP, Stavenhagen JB, Teeling JL. New roles for Fc receptors in neurodegeneration-the impact on Immunotherapy for Alzheimer’s Disease. Frontiers in neuroscience. 2014;8:235. doi: 10.3389/fnins.2014.00235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Eckman EA, Adams SK, Troendle FJ, Stodola BA, Kahn MA, Fauq AH et al. Regulation of steady-state beta-amyloid levels in the brain by neprilysin and endothelin-converting enzyme but not angiotensin-converting enzyme. J Biol Chem. 2006;281(41):30471–8. [DOI] [PubMed] [Google Scholar]
  • 47.Eckman EA, Reed DK, Eckman CB. Degradation of the Alzheimer’s amyloid beta peptide by endothelin-converting enzyme. J Biol Chem. 2001;276(27):24540–8. [DOI] [PubMed] [Google Scholar]
  • 48.Tarasoff-Conway JM, Carare RO, Osorio RS, Glodzik L, Butler T, Fieremans E et al. Clearance systems in the brain-implications for Alzheimer disease. Nat Rev Neurol. 2015;11(8):457–70. doi: 10.1038/nrneurol.2015.119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Leissring MA, Saido TC. Degradation of amyloid-ß protein. In: Selkoe DJ, Mandelkow E, Holtzman DM, editors. The biology of Alzheimer disease. Cold Spring Harbor, New York: Cold Spring Harbor Laboratory Press; 2011. p. 387–404. [Google Scholar]
  • 50.Saido T, Leissring MA. Proteolytic degradation of amyloid beta-protein. Cold Spring Harbor perspectives in medicine. 2012;2(6):a006379. doi: 10.1101/cshperspect.a006379. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Mueller-Steiner S, Zhou Y, Arai H, Roberson ED, Sun B, Chen J et al. Antiamyloidogenic and neuroprotective functions of cathepsin B: implications for Alzheimer’s disease. Neuron. 2006;51(6):703–14. doi:S0896–6273(06)00597–6 [pii] 10.1016/j.neuron.2006.07.027. [DOI] [PubMed] [Google Scholar]
  • 52.Walsh DM, Selkoe DJ. Oligomers on the brain: the emerging role of soluble protein aggregates in neurodegeneration. Protein Pept Lett. 2004;11(3):213–28. [DOI] [PubMed] [Google Scholar]
  • 53.Shankar GM, Leissring MA, Adame A, Sun X, Spooner E, Masliah E et al. Biochemical and immunohistochemical analysis of an Alzheimer’s disease mouse model reveals the presence of multiple cerebral Abeta assembly forms throughout life. Neurobiol Dis. 2009;36(2):293–302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Leissring MA, Farris W, Chang AY, Walsh DM, Wu X, Sun X et al. Enhanced proteolysis of beta-amyloid in APP transgenic mice prevents plaque formation, secondary pathology, and premature death. Neuron. 2003;40(6):1087–93. [DOI] [PubMed] [Google Scholar]
  • 55.Meilandt WJ, Cisse M, Ho K, Wu T, Esposito LA, Scearce-Levie K et al. Neprilysin overexpression inhibits plaque formation but fails to reduce pathogenic Abeta oligomers and associated cognitive deficits in human amyloid precursor protein transgenic mice. J Neurosci. 2009;29(7):1977–86. doi:29/7/1977 [pii] 10.1523/JNEUROSCI.2984-08.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Leissring MA, Turner AJ. Regulation of distinct pools of amyloid beta-protein by multiple cellular proteases. Alzheimer’s research & therapy. 2013;5(4):37. doi: 10.1186/alzrt194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.LaFerla FM, Green KN, Oddo S. Intracellular amyloid-beta in Alzheimer’s disease. Nat Rev Neurosci. 2007;8(7):499–509. [DOI] [PubMed] [Google Scholar]
  • 58.Leissring MA. Abeta degradation-the inside story. Frontiers in aging neuroscience. 2014;6:229. doi: 10.3389/fnagi.2014.00229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Lewis PA, Piper S, Baker M, Onstead L, Murphy MP, Hardy J et al. Expression of BRI-amyloid beta peptide fusion proteins: a novel method for specific high-level expression of amyloid beta peptides. Biochim Biophys Acta. 2001;1537(1):58–62. [DOI] [PubMed] [Google Scholar]
  • 60.Kim J, Onstead L, Randle S, Price R, Smithson L, Zwizinski C et al. Abeta40 inhibits amyloid deposition in vivo. J Neurosci. 2007;27(3):627–33. doi: 10.1523/JNEUROSCI.4849-06.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Kim J, Chakrabarty P, Hanna A, March A, Dickson DW, Borchelt DR et al. Normal cognition in transgenic BRI2-Abeta mice. Mol Neurodegener. 2013;8:15. doi: 10.1186/1750-1326-8-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Leissring MA. The AßCs of Aß-cleaving proteases. J Biol Chem. 2008;283(44):29645–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Matsuoka Y, Saito M, LaFrancois J, Saito M, Gaynor K, Olm V et al. Novel therapeutic approach for the treatment of Alzheimer’s disease by peripheral administration of agents with an affinity to beta-amyloid. J Neurosci. 2003;23(1):29–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Liu Y, Studzinski C, Beckett T, Guan H, Hersh MA, Murphy MP et al. Expression of neprilysin in skeletal muscle reduces amyloid burden in a transgenic mouse model of Alzheimer disease. Mol Ther. 2009;17(8):1381–6. doi:mt2009115 [pii] 10.1038/mt.2009.115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Liu Y, Studzinski C, Beckett T, Murphy MP, Klein RL, Hersh LB. Circulating neprilysin clears brain amyloid. Mol Cell Neurosci. 2010;45(2):101–7. doi:S1044–7431(10)00134-X [pii] 10.1016/j.mcn.2010.05.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Henderson SJ, Andersson C, Narwal R, Janson J, Goldschmidt TJ, Appelkvist P et al. Sustained peripheral depletion of amyloid-beta with a novel form of neprilysin does not affect central levels of amyloid-beta. Brain. 2014;137(Pt 2):553–64. doi: 10.1093/brain/awt308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Walker JR, Pacoma R, Watson J, Ou W, Alves J, Mason DE et al. Enhanced proteolytic clearance of plasma Abeta by peripherally administered neprilysin does not result in reduced levels of brain Abeta in mice. J Neurosci. 2013;33(6):2457–64. doi: 10.1523/JNEUROSCI.3407-12.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Song ES, Juliano MA, Juliano L, Hersh LB. Substrate activation of insulin-degrading enzyme (insulysin). A potential target for drug development. J Biol Chem. 2003;278(50):49789–94. doi: 10.1074/jbc.M308983200. [DOI] [PubMed] [Google Scholar]
  • 69.Abdul-Hay SO, Lane AL, Caulfield TR, Claussin C, Bertrand J, Masson A et al. Optimization of peptide hydroxamate inhibitors of insulin-degrading enzyme reveals marked substrate-selectivity. J Med Chem. 2013;56(6):2246–55. doi: 10.1021/jm301280p. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Marr RA, Rockenstein E, Mukherjee A, Kindy MS, Hersh LB, Gage FH et al. Neprilysin gene transfer reduces human amyloid pathology in transgenic mice. J Neurosci. 2003;23(6):1992–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Spencer B, Marr RA, Rockenstein E, Crews L, Adame A, Potkar R et al. Long-term neprilysin gene transfer is associated with reduced levels of intracellular Abeta and behavioral improvement in APP transgenic mice. BMC Neurosci. 2008;9:109. doi:1471–2202-9–109 [pii] 10.1186/1471-2202-9-109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Tuszynski MH, Thal L, Pay M, Salmon DP, U HS, Bakay R et al. A phase 1 clinical trial of nerve growth factor gene therapy for Alzheimer disease. Nat Med. 2005;11(5):551–5. doi: 10.1038/nm1239. [DOI] [PubMed] [Google Scholar]
  • 73.Tuszynski MH, Yang JH, Barba D, U HS, Bakay RA, Pay MM et al. Nerve Growth Factor Gene Therapy: Activation of Neuronal Responses in Alzheimer Disease. JAMA neurology. 2015;72(10):1139–47. doi: 10.1001/jamaneurol.2015.1807. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Li Y, Wang J, Zhang S, Liu Z. Neprilysin gene transfer: A promising therapeutic approach for Alzheimer’s disease. J Neurosci Res. 2015;93(9):1325–9. doi: 10.1002/jnr.23564. [DOI] [PubMed] [Google Scholar]
  • 75.Barua NU, Miners JS, Bienemann AS, Wyatt MJ, Welser K, Tabor AB et al. Convection-enhanced delivery of neprilysin: a novel amyloid-beta-degrading therapeutic strategy. J Alzheimers Dis. 2012;32(1):43–56. doi: 10.3233/JAD-2012-120658. [DOI] [PubMed] [Google Scholar]
  • 76.Iwata N, Sekiguchi M, Hattori Y, Takahashi A, Asai M, Ji B et al. Global brain delivery of neprilysin gene by intravascular administration of AAV vector in mice. Scientific reports. 2013;3:1472. doi: 10.1038/srep01472. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Spencer BJ, Verma IM. Targeted delivery of proteins across the blood-brain barrier. Proc Natl Acad Sci U S A. 2007;104(18):7594–9. doi: 10.1073/pnas.0702170104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Spencer B, Verma I, Desplats P, Morvinski D, Rockenstein E, Adame A et al. A Neuroprotective Brain-penetrating Endopeptidase Fusion Protein Ameliorates Alzheimer Disease Pathology and Restores Neurogenesis. J Biol Chem. 2014;289(25):17917–31. doi: 10.1074/jbc.M114.557439. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Jacobsen JS, Comery TA, Martone RL, Elokdah H, Crandall DL, Oganesian A et al. Enhanced clearance of Abeta in brain by sustaining the plasmin proteolysis cascade. Proc Natl Acad Sci U S A. 2008;105(25):8754–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Van Nostrand WE, Melchor J, Wagner M, Davis J. Cerebrovascular smooth muscle cell surface fibrillar A beta. Alteration of the proteolytic environment in the cerebral vessel wall. Ann N Y Acad Sci. 2000;903:89–96. [DOI] [PubMed] [Google Scholar]
  • 81.Efanov AM, Barrett DG, Brenner MB, Briggs SL, Delaunois A, Durbin JD et al. A novel glucokinase activator modulates pancreatic islet and hepatocyte function. Endocrinology. 2005;146(9):3696–701. [DOI] [PubMed] [Google Scholar]
  • 82.Cabrol C, Huzarska MA, Dinolfo C, Rodriguez MC, Reinstatler L, Ni J et al. Small-molecule activators of insulin-degrading enzyme discovered through high-throughput compound screening. PLoS ONE. 2009;4(4):e5274. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Bae SJ, Matsunaga Y, Takenaka M, Tanaka Y, Hamazaki Y, Shimizu K et al. Substance P induced preprotachykinin-a mRNA, neutral endopeptidase mRNA and substance P in cultured normal fibroblasts. Int Arch Allergy Immunol. 2002;127(4):316–21. [DOI] [PubMed] [Google Scholar]
  • 84.Mohajeri MH, Kuehnle K, Li H, Poirier R, Tracy J, Nitsch RM. Anti-amyloid activity of neprilysin in plaque-bearing mouse models of Alzheimer’s disease. FEBS Lett. 2004;562(1–3):16–21. [DOI] [PubMed] [Google Scholar]
  • 85.Mohajeri MH, Wollmer MA, Nitsch RM. Abeta 42-induced increase in neprilysin is associated with prevention of amyloid plaque formation in vivo. J Biol Chem. 2002;277(38):35460–5. [DOI] [PubMed] [Google Scholar]
  • 86.Belyaev ND, Kellett KA, Beckett C, Makova NZ, Revett TJ, Nalivaeva NN et al. The transcriptionally active amyloid precursor protein (APP) intracellular domain is preferentially produced from the 695 isoform of APP in a {beta}-secretase-dependent pathway. J Biol Chem. 2010;285(53):41443–54. doi: 10.1074/jbc.M110.141390. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Pardossi-Piquard R, Petit A, Kawarai T, Sunyach C, Alves da Costa C, Vincent B et al. Presenilin-dependent transcriptional control of the Abeta-degrading enzyme neprilysin by intracellular domains of betaAPP and APLP. Neuron. 2005;46(4):541–54. [DOI] [PubMed] [Google Scholar]
  • 88.Huang J, Guan H, Booze RM, Eckman CB, Hersh LB. Estrogen regulates neprilysin activity in rat brain. Neurosci Lett. 2004;367(1):85–7. doi: 10.1016/j.neulet.2004.05.085 S0304394004006822 [pii]. [DOI] [PubMed] [Google Scholar]
  • 89.Lazarov O, Robinson J, Tang YP, Hairston IS, Korade-Mirnics Z, Lee VM et al. Environmental enrichment reduces Abeta levels and amyloid deposition in transgenic mice. Cell. 2005;120(5):701–13. [DOI] [PubMed] [Google Scholar]
  • 90.Saito T, Iwata N, Tsubuki S, Takaki Y, Takano J, Huang SM et al. Somatostatin regulates brain amyloid beta peptide Abeta42 through modulation of proteolytic degradation. Nat Med. 2005;11(4):434–9. doi:nm1206 [pii] 10.1038/nm1206. [DOI] [PubMed] [Google Scholar]
  • 91.Melzig MF, Janka M. Enhancement of neutral endopeptidase activity in SK-N-SH cells by green tea extract. Phytomedicine. 2003;10(6–7):494–8. [DOI] [PubMed] [Google Scholar]
  • 92.Parachikova A, Green KN, Hendrix C, LaFerla FM. Formulation of a medical food cocktail for Alzheimer’s disease: beneficial effects on cognition and neuropathology in a mouse model of the disease. PLoS One. 2010;5(11):e14015. doi: 10.1371/journal.pone.0014015. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES