Abstract
Alzheimer’s disease (AD) is a neurodegenerative disorder of progressive dementia that is characterized by the accumulation of beta-amyloid (Aβ)-containing neuritic plaques and intracellular Tau protein tangles. This distinctive pathology indicates that the protein quality control is compromised in AD. Autophagy functions as a “neuronal housekeeper” that eliminates aberrant protein aggregates by wrapping then into autophagosomes and delivering them to lysosomes for degradation. Several studies have suggested that autophagy deficits in autophagy participate in the accumulation and propagation of misfolded proteins (including Aβ and Tau). In this review, we summarize current knowledge of autophagy in the pathogenesis of AD, as well as some pathways targeting the restoration of autophagy. Moreover, we discuss how these aspects can contribute to the development of disease-modifying therapies in AD.
Keywords: Alzheimer's disease, autophagy, amyloid beta, tau, propagation of amyloid beta and tau, mTOR-dependent pathway, mTOR-independent pathway, autophagy-related interventions
1. Introduction
Alzheimer’s disease (AD) is the most common cause of progressive dementia in the elderly population worldwide. The predominant histological signs of AD are characterized by extracellular amyloid plaques consisting of amyloid-beta (Aβ) and by intracellular neurofibrillary tangles (NFTs) composed of hyperphosphorylated Tau protein. In particular, oligomerization of Aβ peptide in the brain is an early pathophysiological event that occurs a decade or more before symptom onset [1, 2]. In the healthy brain, the amyloid precursor protein (APP) is generated by α-, β- and γ-secretases through either non-amyloidogenic or amyloidogenic pathways, and is assumed to be in equilibrium. Meanwhile, in AD brain, this balance breaks down and the amyloidogenic pathway is dominant for the cleavage of APP, giving rise to peptide fragments known as Aβ [3, 4]. Once formed, Aβ (and in particular Aβ42) abnormally accumulates, resulting in the formation of Aβ oligomers, cerebral amyloid plaques, neurodegeneration, and ultimately brain atrophy. Tau, is typically localized to axons where it binds and stabilizes microtubules based on its phosphorylation status. In AD, there are high levels of intraneuronal hyperphosphorylated tau and aberrant tau misfolding due to hyperphosphorylation and dissociation from microtubules [5]. This instability of microtubules leads to the severe disruption of axonal transport, which results in neuronal death. Following neuronal death, Tau filaments are aggregated in the extracellular space, and are the major components in the formation of the other types of AD-associated neuropathological lesion, namely, NFTs. Increasing evidence suggest that pathological Tau abnormality is as important as amyloid plaques in AD development [6] (Fig. 1). Besides aggregation of these two misfolded proteins, Tau and Aβ propagation is responsible for the progression of AD. In 2006, the progressive spread of Aβ deposition was first detected in APP transgenic mice [7]. Soon after, prion-like mechanisms of Tau pathology were verified [8]. Since then, multiple studies suggest that Aβ deposits and Tau assemblies can “seed” the formation of aggregates, followed by their spreading to other cells [9-12].
Fig. (1).
Amyloid deposits and neurofibrillary tangles (NFTs) are the two main neuropathological lesions described in patients with Alzheimer disease (AD). (A higher resolution / colour version of this figure is available in the electronic copy of the article).
Autophagy is the key intracellular degradation pathway for eliminating aberrant proteins and promoting survival and suppressing programmed cell death [13]. In case of autophagy, intracellular waste is enwrapped by double-membraned vesicles termed as phagophores and degraded by fusion with lysosomes [14, 15]. Under normal conditions, autophagy is constitutively active and eliminates the cytotoxic effect of damaged proteins and organelles to maintain intracellular homeostasis [16, 17]. During the progression of AD, autophagy is aberrant. Dysfunctional autophagy resulting in the accumulation of mutated toxic proteins (i.e., Aβ and Tau), which causes an increase in oxidative stress and neuronal death [18]. Besides, autophagy also interferes with propagation of both misfolded proteins. Exosomes are recognized as the cargo-laden vesicles that help amyloid transmission. During autophagic dysfunction, multivesicular bodies release intraluminal vesicles (ILVs) containing amyloids as exosomes, thereby decreasing the proteotoxic stress in the releasing cells, yet propagating protein aggregates to neighboring cells [19]. Furthermore, the pathological changes of AD also lead to defective autophagy. Persistent accumulation of Aβ-induced aberrant autophagy and decreased autophagosome‐lysosome fusion constitute a vicious loop that worsens AD [20].
In this review, we summarize the current knowledge on the relevance of autophagy dysfunction to AD-related pathology. Molecular targets for autophagy modulation and potentially therapeutic drugs mediating autophagic flux disruption in AD will also be reviewed. We hope our work will foster a hopeful therapeutic platform for Alzheimer's disease.
2. Autophagy machinery
Autophagy is a finely organized phenomenon through which eukaryotic cells can digest overabundant organelles, macromolecules and proteins and recycle their breakdown products [21, 22]. Simply, this degradation pathway is initiated by the fusion of vesicles to form a phagophore, a cup-shaped double-membrane, enveloping small fragments of the cytoplasm and organelles. By elongating, phagophores gradually sequester cargo until they form a double-membrane bound vesicles termed as autophagosomes. By means of cytoskeletal microtubule systems, autophagosomes slide along and fuse with different vesicles, and ultimately with lysosomes. Finally, a single-membraned autolysosome is formed, which allows degradation of the autophagosome contents by lysosomal hydrolases [23, 24].
Autophagy induction is finely regulated by two major complexes: the Unc-51 like autophagy activating kinase (ULK1) complex (consisting of ULK1, autophagy-related protein [ATG]-13, ATG101, and focal adhesion kinase family interacting protein of 200 kDa [FIP200]) and class III phosphatidylinositol 3-kinase (PI3K)/Beclin-1 complex (consisting of vacuolar protein sorting [VPS]-34, VPS15, ATG6, and ATG14) [24, 25], which are sensors for intracellular homeostasis. The ULK1 complex is mainly regulated by the mammalian target of rapamycin complex 1 (mTORC1) and 5′ AMP-activated protein kinase (AMPK) pathway, which acts as an autophagy blocker and activator [26]. Moreover, p53 is also suggested to transcriptionally regulate ULK1 under oxidative stress conditions [27], as elaborated below. Activated ULK1 recruits PI3K and VPS34, which induces accumulation of phosphatidylinositol-3-phosphate (PI3P) in phagophores and drives autophagosome nucleation [28, 29]. In this step, the Beclin-1 interactome works as a precise regulator. Under normal conditions, Beclin-1 binds to B-cell lymphoma 2 (Bcl-2) to suppress the formation of PI3K complex, leading to the inhibition of autophagy. Meanwhile, under stress conditions, Beclin-1 interacts with VPS34 and triggers the activation of PI3K thus stimulating autophagocytosis [30] (Fig. 2).
Fig. (2).
Schematic overview of the general autophagy pathways. (A higher resolution / colour version of this figure is available in the electronic copy of the article).
3. Biological mechanisms linking autophagy and AD
Neurons are characterized by the complex axonal and dendritic structures, which required intense transport and efficient proteostasis to accommodate a dynamic microenvironment [31]. Unlike other eukaryotes, neurons are post-mitotic and are unable to dilute toxic substances by mitosis. Hence, the quality of cytoplasmic and protein/organelles clearance is orchestrated to a large extent by autophagy. However, AD weakens autophagic function, eventually resulting in abnormal accumulation of autophagic substrates and lowering the basal autophagic flux. Nixon et al., [26] examined post-mortem AD brain and found dystrophic neurites to contain immature autophagosomes. Accumulation of autophagosomes is more likely to arise from impaired clearance rather than the induction of autophagy itself. However, autophagy might also regulate AD-related pathology. Nilsson et al., observed that the offspring of autophagy-deficient mice showed greater intracellular aggregation of Aβ together with amyloidosis and memory impairment than mice with functional autophagy [32]. Depleting a key autophagy-related gene, namely Becn1 (which encodes Beclin-1), suppresses induction of autophagy and leads to AD in the mice model [33]. Moreover, the regulation of autophagy in brain is region- and tissue-specific [24]. In AD, the autophagic mechanism is differentially affected within the cortex, hypothalamus, midbrain, and cerebellum. In addition to these regional differences, neurons are not equally affected within the same region. Hence, because of the inherent reliance on autophagy, specific regions or neurons may be more prone to protein aggregation, leading to a specific clinical phenotype. Altogether, these findings clearly show that the role of the autophagic system is not only a causative or protective factor but also a consequence of AD.
3.1. Autophagy is a Key Regulator of Aβ Metabolism
First, degradation is considered the first function of autophagy. Indeed, autophagy is considered to be another main Aβ eliminating pathway besides Aβ-degrading enzyme [34], which is especially prevalent in the AD brains. Many studies have shown that autophagy works on reducing extracellular Aβ plaque burden as well as peptide Aβ fragments inside brain cells [32, 35, 36]. Microglia are the resident brain macrophages that constantly survey the neural environment for pathogens and apoptotic cells. In the vicinity of extracellular Aβ plaques, microglia assembled and attempt to clear the pathological deposits of Aβ through phagocytosis. After internalization by microglia, Aβ may leak out of the endolysosomal system into the cytosol, resulting in cellular damage [35]. This phenomenon requires the autophagic machinery for Aβ fibrils degeneration. Moreover, Aβ-induced inflammatory mediators released from microglia damage neurons, and this inflammatory response is controlled by microglial autophagy. In a recent study, Aβ-induced NLRP3 inflammasome activation is aggravated by impaired autophagy [35]. Second, beyond conventional Aβ degradation, a regulation of Aβ secretion and plaque formation is another major function of autophagy in AD [32]. Because Aβ is mainly located on membranes, the presence of Aβ has also been found in autophagic vesicles (aside from the Golgi and endosomes) in patients with AD [37]. This finding indicates that the accumulation of immature autophagic vesicles may be the origin of Aβ production under pathological conditions. In autophagy-deficient mice, Aβ production is decreased by 90%. However, recovery of autophagy reverses Aβ secretion to normal levels [37]. These results do not mean autophagy is an accelerator of AD progress especially for Aβ pathology. On the contrary, this reduction of Aβ plaque load leads to Aβ aberrant intraneuronal accumulation which results in aggravating the damage to neurons [32]. Third, genes essential for autophagy are also implicated in AD pathology. Inhibition of Beclin-1, an adaptor protein that manipulates several autophagy steps, can increase intracellular and extracellular Aβ deposition [38]. Another key regulator of autophagy, presenilin-1 (PS1) participates in the formation of γ-secretase complex. PS1 has been identified as a causative gene for early-onset AD (also known as familial AD) [39]. As described earlier, the role of PS1 in lysosomal-dependent proteolysis is directly relevant to the mechanism by which PS1 mutations contribute to the onset of AD [40]. In addition, the apolipoprotein E gene (ApoE4) is also widely accepted as a major risk factor for AD. Studies from Neuro-2a cells and transgenic mice have shown that ApoE4 potentiates lysosomal leakage and apoptosis induced by Aβ peptides [41, 42].
3.2. Autophagy Contributes to the Clearance of Tau
Tau is a microtubule-associated protein (MAPT) that polymerizes tubulin into microtubules, and consequently participates in maintaining the complex neuronal cell microarchitecture, including microtubule assembly and stabilization, particularly in the axons [43]. When tau is monomeric, the ubiquitin proteasome system occupies a dominant position. However, when tau is oligomeric or in aggregate forms, autophagy is depended upon for degradation [40]. In 1996, Bednarski reported for the first time that a lysosomal protease can degrade Tau in cultured hippocampal slices, highlighting autophagy in the pathogenesis of Tau metabolism [44]. Since then, it has been shown that reduction of phosphorylated (p)-tau/NFTs is accompanied by improved neuronal survival and function, which requires lysosomal activity both in vitro and in vivo [45]. Ammonium chloride is an inhibitor of autophagy processes that delays the clearance of Tau and promotes some high molecular weight Tau formation. Cathepsin D (a lysosomal protease), rapamycin (an autophagy inducer), and trehalose (an activator of autophagy) can also help degrade Tau and reduce the level of Tau aggregates in Drosophila or in the brains of human Tauopathy model mice [46]. Furthermore, various kinases are included in this process in vivo. Among them, glycogen synthase kinase-3β (GSK-3β), an autophagy upstream kinase, occupies an important status. GSK- 3β is regarded as the most documented Tau kinase for its upregulation, which modulates abnormal hyperphosphorylation of Tau in AD [47]. Furthermore, as a chaperone protein, clusterin participates in autophagosome biogenesis. In patients with AD, tau levels in cerebrospinal fluid were reportedly dysregulated by the AD-associated clusterin polymorphism, rs11136000 [48].
3.3. Autophagy may Halt the Propagation of Aβ and Tau
Besides gradual accumulation of Aβ and Tau brain pathology, the prion-like spreading of pathological proteins in the central nervous system is also accompanied by progression of AD [49]. Accordingly, studies have demonstrated that exogenous inoculation of Aβ-containing brain extracts spreads Aβ to adjacent regions [7, 50, 51]. Similarly, Tau transmission from microglia to neurons has been verified in cellular and animal models was verified [52]. Currently, exosomes are speculated to be responsible for the propagation of Aβ and Tau. Immunoelectron microscopy of purified exosomes from AD brain sections showed that Aβ localizes to exosomes [53]. Further, inhibition of exosomes synthesis halts neuron-to-neuron Tau transmission [52]. In 2015, Edward et al. first identified autolysosomal proteins, such as cathepsin-D and lysosomal-associated membrane protein 1, in neutrally-derived blood exosomes from patients with AD [10, 12], thereby bolstering the interplay between autophagy and exosomal pathways. Furthermore, in primary cortical neurons and Neuro-2a cells, VPS34 inhibition increased secretion of extracellular vesicle-associated APP C-terminal fragments. This supports the hypothesis that endolysosomal dysfunction is the mechanism driving secretion of these fragments from exosomes. Thus, the considerable influence of autophagy in exosomal spread of pathological proteins is crucial for providing new opportunities for perturbing AD progression. However, whether fusion of multivesicular bodies with autophagic vacuoles results in exosome release, or alternatively, exosomes are the production of lysosome degradation or structural differences between autophagic vacuoles and exosomes, still needs further investigation.
4. Activation of Autophagy as a Therapeutic Strategy for AD
As we discussed above, the activation of autophagy may serve as a therapeutic strategy for AD. Hence, we highlight several possible opportunities for therapeutic intervention, namely, mammalian kinase target of rapamycin (mTOR)-dependent and mTOR-independent. These approaches aim to restore autophagy function and have proven efficacious in vitro or in vivo, and with potential for translating to clinical application (Fig. 3).
Fig. (3).
Overview of the regulation of autophagy and potential drug targets. (A higher resolution / colour version of this figure is available in the electronic copy of the article).
5. Key pathways regulating autophagy activity in AD
It has been reported that mTOR serves as a brake on autophagy [54], while high mTOR activity was recently described as a new risk factor for AD [55, 56]. Accordingly, suppression of mTOR enhances Aβ clearance, ameliorates tau pathology, and improves cognitive function in cellular and transgenic models of AD [57-60]. Clearly, there is a direct link between AD pathology and the mTOR-dependent autophagy. Further, mTOR coordinates or interacts with the upstream signaling components, including PI3K/AKT, AMPK, GSK-3, and insulin/insulin-like growth factor 1 (IGF-1), and downstream components, including transcription factor EB (TFEB), nuclear receptor binding factor 2 (NRBF2), and p62 [61-64]. Insight into these complex pathways may provide a comprehensive means to counteract AD progression.
5.1. mTOR-dependent Pathway
5.1.1. PI3K/AKT Signaling
During brain development, the PI3K/AKT pathway plays an important role in controlling neurotransmission, establishing neuronal polarity, and in maintaining balance in neuronal survival/death [65, 66]. In the brains of healthy individuals, phosphoinositide 3 kinase (PI3K) activates mTORC1 via AKT by phosphorylating the tuberous sclerosis complex (TSC2)-2, which in turn inhibits function of the TSC1/TSC2 complex, negatively regulates conversion of Ras homolog enriched in brain (Rheb) [67]. However, with human aging and AD, the PI3K/AKT pathway is disrupted and consistently activated leading to Aβ aggregation and cognitive decline. In the brain of Down’s syndrome patients with AD-like pathology, Perluigi, et al., demonstrated a strong association between aberrant activation of PI3K/AKT pathway and decrement in the autophagic flux (low ratio of microtubule-associated protein 1A/1B-light chain 3 [LC3]-II/LC3-I) via phosphorylation of the ULK1-ATG13-FIP200 complex [68]. Phosphatase and tensin homolog (PTEN), which downregulates PI3K/AKT signaling pathway, is altered in the brain from patients with AD [69-71]. Upregulation of PTEN can relieve Aβ-induced neurotoxicity [72] and decrease the phosphorylation state of Tau [73].
5.1.2. GSK3 Signaling
Long-term inhibition of GSK3 kinases increases autophagic flux by downregulating mTORC1 via phosphorylation of both ribosomal protein S6 kinase beta-1 and ULK1, which reduces the interaction between mTORC1 and raptor [74]. In brain samples from individuals with mild cognitive impairment and late-onset AD, p-GSK3 (the active form of GSK3), is demonstrably reduced compared with controls without dementia. This further suggests that abnormal GSK-3 activity is a pathogenic event in both early- and late-stage AD [75]. Early in the amyloid pathology, GSK3 isozymes become activated by increased tyrosine phosphorylation in the hippocampus of old APP transgene mice [76]. Similarly, APP knockout mice show a dramatic decrease in GSK-3 activity [75]. As GSK-3 accelerated Aβ production by escalating the activity of beta-secretase 1(BACE-1) [77], it implies a feedback loop of GSK-3/Aβ generation. GSK3 exists as two isozymes: GSK3α and GSK3β. GSK3β is identified as the major kinase in Tauopathy as well as a major link from amyloid-to-Tau pathology [76]. Activated GSK3β targets hyperphosphorylated Tau protein, which impairs the Tau-microtubule interaction, leading to eventual aggregation into tangles. In hippocampal pyramidal neurons of aged mice, GSK-3 inhibition not only stimulates autophagy activation but also leads to the compromised autophagy resolution. This occurs through GSK3-dependent regulation of lysosomal biogenesis mediated by the TFEB, and in turn, decreases lysosomal proteolysis [78].
5.1.3. AMPK Signaling
As stated earlier, the ULK1-ATG13-ATG101-FIP200 complex is a critical initiator of autophagy and its activity is mainly regulated by the combination of AMPK [79, 80]. Normally, active mTORC1 phosphorylates ULK1 on Ser 757, thereby weakening autophagy [79]. Upon cellular stresses, AMPK interacts with and phosphorylates ULK1 on Ser 317 and Ser 777, leading to the dissociation of the ULK1-mTORC1 complex, translocation of ULK to the pre-autophagosomal membrane and eventually induction of autophagy [79]. Alternatively, AMPK may inhibit mTORC1 activation by directly phosphorylating raptor on two conserved serine residues (Ser722 and Ser792) [81]. Lately, several new findings have suggested that ULk1 provides potential negative feedback for AMPK activity by phosphorylating all three subunits of AMPK (AMPK-α, β and -γ) [25, 82]. Debate on whether activation of AMPK is neuroprotective or neurotoxic is long-lasting. Molecular biological investigations have shown that AMPK is involved in regulation of Aβ levels.Novel synthetic compounds (RSVA series) targeting the activation of AMPK were identified as promising lead molecules facilitating Aβ clearance [83]. However, Vingtdeux et al. demonstrated that activated AMPK (p-AMPK) accumulated in AD brain in structures that were similar to NFTs, according to immunohistochemical staining. This supports a role of AMPK in preceding tau aggregation and thus triggering AD progression [84].
5.1.4. IGF-1 Signaling
IGF belongs to a class of neurotrophic factors that classically regulate neuronal growth and neuronal survival [85]. In 2009, Mona and colleagues observed that IGF-1 regulated autophagy in Purkinje neurons by accelerating basal autophagosome-lysosome fusion rates in an mTOR-dependent manner [86]. Furthermore, in rat embryonic cortical neurons, an inhibitory effect of IGF-1 on autophagy was demonstrated by binding with its receptor, IGF-1R, together with subsequent activation of the PI3K/AKT/mTOR pathway [87, 88]. This also suggests that IGF-1 treatment may decrease Aβ generation by reducing APP phosphorylation and promoting memory deficits. This result is consistent with a previous study showing that subcutaneous administration of IGF-1 to aged rats decreased Aβ levels in brain parenchyma to levels observed in young rats [89]. For Tauopathy, it has been shown that IGF-1 gene inactivation reduces aberrant tau phosphorylation and potentiates NFT formation [90].
5.1.5. mTOR/TFEB Signaling
TFEB was identified as a positive regulator of autophagy by inducing a broad transcriptional program and controlling autophagosome-lysosome fusion [91]. Inactive TFEB is normally sequestered in the cytoplasm by phosphorylation, while dephosphorylated TFEB translocates to the nucleus and induces activation of transcriptional downstream targets [92, 93]. This event is mediated by binding to the key negative autophagy regulator, MTORC1, which contains coordinated lysosomal expression and regulation (CLEAR) element [94]. However, recently Diego et al., [95]. detected overexpression of calcineurin-induced TFEB nuclear translocation in normally fed cells, providing a novel mTORC1-independent regulator for TFEB. For amyloid pathology, TFEB enhanced lysosomal biogenesis in both astrocytes and neurons, participating in the progression of Aβ clearance and Aβ generation, respectively [96, 97]. Further evidence showed that this effect of TFEB was diminished with inhibition of GSK3β activity in both PS1-deficient mouse models and neural stem cell models [98]. Regarding Tau, exogenous TFEB expression in the rTg4510 mouse brains and T40PL cells (both Tauopathy models) increased misfolded Tau proteins clearance [5]. The authors hypothesized that an AKT-mTOR-TFEB feedback regulatory loop is involved in the p-Tau clearance by inducing autophagy.
5.1.6. mTOR/NRBF2 Signaling
NRBF2 is a novel component of the PtdIns3K complex that is associated with ATG14 in mammalian cells [99]. Ma et al. reported that NRBF2 functions as a fine-tuning autophagy regulator. Under normal conditions, NRBF2 is phosphorylated by MTORC1 and preferentially binds a non-autophagic form of PtdIns3K, leading to autophagy inhibition. When MTORC1 is inhibited, NRBF2 becomes unphosphorylated. This form of NRBF2 binds ATG14- Beclin-1, increasing autophagic PtdIns3K complex assembly, stimulating phosphatidylinositol 3-phosphate production, promoting ULK1 association and leading to the activation of autophagy [100]. Recently, it was found that NRBF2 was downregulated in the hippocampus of 5xFAD mice and NRBF2 overexpression reduced Aβ1-42 levels by modulating autophagic progression [101].
5.1.7. mTOR/p62 Signaling
The landmark discovery of p62/SQSTM1 as the first mammalian selective autophagy receptor defined a new family of autophagy-related proteins. p62 is a multifunctional protein rich in domains, among which, the LC3‐interacting motif (LIR) and ubiquitin-binding domain (UBA) mainly controlling autophagy uptake. Through its LC3‐interacting motif, p62 binds to LC3 to facilitate selective substrate recognition [102, 103]. Through the ubiquitin-binding domain, p62 binds to polyubiquitinated proteins (including Tau), and shuttles them to autophagosomes for degradation [104, 105]. Further, p62 also interferes with the protein interaction between BCL-2 and Beclin-1 during the autophagy process [30]. For mitophagy, p62 balances mitochondrial quality control and recruits damaged mitochondria to the phagophore by connecting with Parkin [106]. By analyzing AD patients’ brain, Du et al., [107] found that p62 expression is significantly reduced in the frontal cortex. To dissect the role of p62 in reducing Aβ levels, Antonella and colleagues generated APP/PS1 mice with an adeno-associated virus expressing p62. They found better spatial learning performance in APP/PS1–p62 mice compared with APP/PS1-GFP mice. This study also indicated that p62 decreases Aβ levels by facilitating the delivery of Aβ to lysosomes for degradation [108]. Alternatively, p62 knockout mice showed age-dependent accumulation of NFTs and synaptic deficits [109]. Thus, decreased p62 levels directly lead to low autophagy levels, which may accelerate Tau and Aβ protein aggregation resulting in AD progression. Correlation between mTOR activity and p62 is still controversial. It has been reported that p62 acts as a downstream target of mTOR in the cortical neurons [110] and that decreased mTOR precedes or coincides with a decrease in p62 [111]. Contradictorily, studies also revealed that p62 directly binds to mTOR as an indispensable, activating component for mTORC1 [112, 113]. The reason for this discrepancy is still not known and requires further investigation.
5.2. mTOR-independent Pathways
Although the canonical mTOR dependent autophagy pathway has been intensively studied, autophagy can be initiated by mTOR-independent pathways and mediated through non-canonical signaling events as well. Here we will highlight several alternative pathways in AD.
5.2.1. SIRT1
The mammalian silent information regulator 2 homolog-1 (SIRT1) belongs to the family of NAD-dependent protein deacetylases, and has a crucial function in governing longevity [114]. Furthermore, therapeutic upregulation of SIRT1 provides an opportunity for the amelioration of Aβ neuropathology through inhibition of amyloidogenesis [115]. In 2008, Lee et al., [116] demonstrated a role for SIRT1 in regulation of autophagy. In particular, transient increased expression of SIRT1 was sufficient to stimulate basal rates of autophagy. SIRT1 governs the formation of autophagic vacuoles by deacetylation of essential autophagy proteins, such as Atg5, Atg7 and LC3 [116]. In addition, SIRT1 also boosted autophagy via AMPK activation, and this effect was abolished by the AMPK inhibitor, compound C [117]. In APP/PS1 mice, SIRT1 overexpression reduced Aβ production and Aβ plaques, whereas deleting SIRT1 abolished this protective effect [118]. Another study indicated that incubation of Aβ1-40 and Aβ1-42 fragments with an activator of SIRT1, resveratrol, reduced the length and number of these fibrils [119]. In Tau transgene mice, SIRT1 deletion increased Tau mislocalization postsynaptically and Tau propagation from the injection site to the contralateral hippocampus [120]. Thus, SIRT1 deficiency is now regarded as a causative in cognitive decline [121].
5.2.2. p53
The transcriptional regulator p53 safeguards cells against stress and dysfunction, by controlling cell proliferation and apoptosis. p53 is also cast as a pro-autophagic factor that is directly driven by a specific subcellular localization. Briefly, p53 positively modulates autophagy in the nucleus, whereas cytoplasmic p53 inhibits the induction of autophagy conversely [122]. Many p53 target genes (including PTEN, TSC2 and AMPK) block mTOR signaling, leading to the inhibition of autophagy [123]. Involvement of p53 in the control of mitophagy has also been reported [124]. In AD, increased p53 levels were detected in various regions of patient brains compared with healthy individuals [125, 126]. Likewise, γ-secretase complex (composed of PS1 or PS2, nicastrin, gamma-secretase subunit Aph-1, and presenilin enhancer 2)-mediated p53-dependent neuronal death. In turn, p53 may enhance expression of PS-1 and PS-2, which results in cross-talk between p53 and Aβ production [127]. For Tauopathy, studies revealed that sustained expression of p53 promoted Tau phosphorylation which precipitates formation of NFTs [128].
5.2.3. Beclin-1
Whereas mTOR negatively regulates initial autophagosome formation, Beclin-1 regulates both autophagosome formation and autolysosome fusion. By binding with VPS34, Beclin-1 triggers formation of endosomes and autophagosomes, which stimulates the onset of autophagy. Upon binding with BCL-2, Beclin-1 dissociates from Vps34 and inhibits autophagic cell death [129]. Beclin-1 also interacts with PTEN-induced kinase 1 (PINK1) in mitochondria, which can stimulate the clearance of defective mitochondria [130]. With a key role in modulating autophagy, the Beclin-1 interactome is an important regulator of AD pathology. Patients with AD express lower levels of Beclin-1 than age-matched controls [38]. Pickford et al., [38] cross-bred Beclin-1-/- mice with APP transgenic mice, and demonstrated robust deposition of Aβ aggregates. Subsequently, Altea et al., showed that genetically disrupting Beclin-1/BCL-2 binding in 5xFAD mice hyperactivates autophagy and leads to sequestration of amyloid oligomers [33]. Beclin-1 is also reportedly present in tau-positive dystrophic neurites, where it interacts with the PINK/Parkin pathway to facilitate autophagosome maturation and enhance tau degradation [131].
5.2.4. PPARα
The nuclear receptor, peroxisome proliferator-activated receptor alpha (PPARα) is characterized as a pivotal regulator of energy metabolism and peroxisomal function [132]. In response to PPARα activation, normal suppression of autophagy is reversed and the autophagic lipid degradation is induced. Specifically, levels of LC3-II and other indicators of autophagy are upregulated and associated with decrease in inhibitory phosphorylation of ULK1 [79, 133]. Further, activation of PPARα also leads to the increased transcription of TFEB and stimulates lysosomal biogenesis in brain cells [134]. Recently, Luo et al., demonstrated that enhanced PPARα expression protects against amyloid pathology and cognitive decline though its role in activating autophagy and lysosomal pathway [135]. In accordance with this, Chandra S et al., [136] further delineated that cerebral Aβ plaques ware markedly reduced in 5xFAD mice via upregulation of TFEB in a PPARα-dependent fashion.
5.2.5. NRF2
Next to the TFEB and FOXO family of transcription factors, nuclear factor erythroid-2 related factor 2 (NRF2) also participates in the regulation of autophagy-related genes, including ULK, ATG7, and ATG5 [137]. Once activated, NRF2 dissociates from Kelch-like ECH-associated protein 1 (KEAP1) and translocates from cytoplasm to the nucleus, where it coordinates a battery of antioxidant response element (ARE) dependent genes [138]. Dissociated KEAP1 interacts with both LC3-II and p62, facilitating the formation of autophagosomes and relieving NRF2 from inhibition. In aged APP/PS1 mice, NRF2 expression significantly improved learning and memory retention [139]. To further determine the relevance of NRF2 and amyloidopathy, a mouse model (namely AT-NRF2-KO) was generated lacking an obvious amyloid load and NRF2-expression. As early as 6 months, the AT-NRF2 KO mice precede the aggregation of Aβ and Tau filaments with reduced hippocampal long-term potentiation [140]. In human neurons suffering from proteotoxic APP/Aβ insults, an increased nuclear NRF2 expression and the upregulation of the autophagy proteins P62 and ATG7 indicated that NRF2 precludes Aβ formation by activating an autophagy defensive program [137]. Another autophagic receptor, nuclear domain 10 protein (NDP52), also appears to be induced by NRF2 and directly degrades phosphorylated Tau due to increased autophagic clearance [141, 142].
5.2.6. Lysosomal Calcium Signaling
Transient receptor potential mucolipin-1 (TRPML1; also named MCOLN1) is a cation-permeable channel predominantly localized to late endosomal/lysosomal membranes, which regulates lysosomal calcium release during lysosomal fusion with other membranes [143]. Beyond the role of TRPML1 in endolysosomal trafficking, new findings show that TRPML1 facilitated nuclear translocation of TFEB and stimulated transcription of its target genes. In cells silenced for TRPML1, the number of PI(3)P-positive vesicles and the levels of autophagic markers (LC3, P62 and Beclin-1) is reduced indicating the impairment of autophagosome formation [24, 95]. Moreover, TRPML1 also inhibits inhibit AD progression. For example, in primary neurons, activation of TRPML1 promotes the clearance of intraneuronal Aβ from lysosomal compartments [144]. The mucolipidosis IV mouse model (i.e., TRPML1−/− mice) display progressive behavioral and locomotor deficits and cognitive impairment [145]. In APP/PS1 transgenes mice, TRPML1 overexpression markedly improved the recognition index with a significant increase of autophagic markers (LC3, P62 and Beclin-1) and concurrent with the inhibition of AMPK/mTOR pathway, indicating TRPML1 decreases protein aggregates by regulating endosomal–lysosomal pathway [146].
5.3. PINK1/Parkin-dependent Mitophagy
Accumulation of damaged mitochondria in axons and at synapses is a hallmark of age-related diseases, including AD. Electron microscopy images of postmortem human hippocampus samples from patients with AD showed altered mitochondrial morphology, while Basal levels of mitophagy being 30%-50% lower than normal controls [147]. Parkin mediates the removal of damaged mitochondria [148], and PINK-Parkin pathway has emerged as a key regulatory pathway of mitophagy [149]. Under physiological conditions, PINK1 is continuously imported into mitochondria for cleavage and degradation [150, 151]. However, in AD, PINK1 accumulates on the outer membrane of mitochondrial and phosphorylates Parkin at Ser65 (which is necessary for its activation) [152-154]. Thus, PINK1 can act as a tag for mitochondrial injury. Activated Parkin prevents damaged mitochondria from integrating with other healthy mitochondria by ubiquitinating mitofusin-2 protein present in the mitochondrial membrane. Consequently, the damaged mitochondria are eventually degraded in the autophagic pathway via binding to LC3 through P62 [155]. In accordance with these findings, PINK1 overexpression attenuated Aβ accumulation by inducing more abundant expression of autophagy proteins, such as LC3 and NDP52, in APP/PINK1 mice [156]. Moreover, Parkin was selectively recruited to the depolarized mitochondria, which led to elevated levels of LC3-II and p62, resulting in increased post-translational APP expression [157, 158]. All this suggests that enhancement of PINK1-Parkin pathway can improve the outcome of AD progression by facilitating mitophagy functions. Recently, Fang et al. [147] systematically demonstrated activation of mitophagy improving memory functions, ameliorating Aβ pathology, and promoting phagocytic activity of microglia in transgene AD mice. This group also showed restoration of mitophagy by PINK1 overexpression lowered intraneuronal tau levels and mitigated NLRP3/caspase-1-dependent neuroinflammation.
6. Autophagy-related Interventions
Based on the aforementioned information, the most effective way to treat AD is to prevent the aggregation of Aβ and Tau proteins. Autophagy is a well-recognized scavenger that can degrade these aged and malfunctioning proteins. Thus, autophagy-stimulating interventions may have promising therapeutic targets for AD treatment in clinical trials. Here, we shortly discuss interventions under investigation (Table 1).
Table 1.
Autophagy-stimulating compounds and life style interventions which are currently under-study for AD treatment.
| Compound | Therapeutic Class | Mechanism of Autophagy | Refs. |
|---|---|---|---|
| Rapamycin | Immunosuppressant | Inhibition of mTORC1 | [61, 160, 161, 163] |
| Ibuprofen | Nonsteroidal anti-inflammatory drug | inhibition of GSK3β | [166] |
| Aspirin | Nonsteroidal anti-inflammatory drug | ● PPAR-mediated upregulation of TFEB ● Regulation of mitophagy |
[167] [168] |
| Lithium | Anti-psychiatric | Inhibition of GSK-3β | [171, 172] |
| Memantine | Non-competitive NMDA receptor antagonist | ● Promotion of mitophagy ● Inhibition PI3K/Akt/mTOR pathway |
[174] [175] |
| Melatonin | Neurohormone | ● Activation of SIRT1 | [178, 179] |
| Metformin | Antidiabetic | ● Activation of AMPK ● Activation of SIRT1 ● Facilitating Parkin-mediated mitophagy |
[186] [181] [183] |
| Vitamin D | Essential Vitamin | ● Activation of AMPK ● Calcium signaling ● Disruption of Beclin-1 |
[194] [194] [193] |
| Trehalose | Natural disaccharide | Unknown | [195, 196, 197] |
| Resveratrol | Neutraceutical | ● Activation of SIRT1 ● Activation of AMPK ● Inhibition of mTORC1 |
[118, 119, 179] [199] [198] |
| Quercetin | Neutraceutical | ● Activation of Nrf2 ● Inhibition of GSK3β |
[204] [205] |
| Ginsenosides | Neutraceutical | ● Activation of Nrf2 ● Inhibition of mTOR ● Inhibition of GSK3β |
[208] [207, 209] [210] |
| Curcumin | Neutraceutical | ● inhibition of PI3K/Akt ● mTOR-independent upregulation of TFEB |
[213] [215] |
| Calorie restriction/Fasting | Lifestyle interventions | ● Activation of SIRT1 ● Activation of IGF-1 |
[115, 224] [223] |
| Physical Activity | Lifestyle interventions | ● Activation of AMPK ● Upregulation of p62 ● Upregulation of BECN1 |
[230] [228] [229] |
6.1. Autophagy Modulating Drugs
6.1.1. Rapamycin
Rapamycin (also known as Sirolimus) is an mTOR inhibitor, and was the first drug that has been experimentally proven to slow down the progress of aging [159]. This anti-aging ability of rapamycin is ascribed to its suppression of the activation of mTOR-dependent autophagy flux. In neuronal cultures, toxicity of different amyloidogenic peptides can be counteracted by rapamycin [61, 160]. In transgenic mouse models of AD, rapamycin prevented learning and memory deficits and reduced amyloid plaque burden [161]. However, a previous publication suggested that rapamycin is only useful for patients in the very early stages of AD [162]. Intervention with rapamycin at later points would add “fuel to this fire” by exacerbating existing lysosomal problems. Although treatment with rapamycin increased the generation rate of lysosomal-system cargo, during the last period of AD, this would result in accumulation of lysosomal vesicles and further promotion of transcellular seeding of Aβ and Tau by exosomes. In addition, clinical application of rapamycin should also consider adverse effects due to mTOR inhibition because cellular functions besides autophagy are potentially disturbed [163]. Common adverse reactions of rapamycin include respiratory and urinary tract infections, gastrointestinal pain, thrombocytopenia, and dyslipidemia. Temsirolimus is an esterified rapamycin derivative. Because of its tolerable side-effects compared with rapamycin, temsirolimus is under investigation as a potential therapeutic extension to long-term treatment of AD by enhancing autophagy activity [164].
6.1.2. Nonsteroidal Anti-inflammatory Drugs (NSAIDs)
Based on a large health care database, scientists found that participants with a long-term use of NSAIDs were at lower-than-expected risk of AD of >5 years due to suppressed formation of Aβ1-42 [165]. Among individual NSAIDs, ibuprofen and aspirin showed the most marked neuroprotective effect. Ibuprofen reduced Tau pathology by inhibiting of the GSK3β pathway [166]. Meanwhile, aspirin exhibited amyloid-lowering effects by stimulating lysosomal biogenesis via PPAR-mediated upregulation of TFEB [167]. Acetylation is a key mechanism for regulating mitophagy. Appropriately, aspirin inhibited oligomerization of amyloid peptides and subsequent formation of insoluble aggregates by disrupting mitochondrial protein acetylation, and further strengthening the potential therapeutic role of aspirin in AD [168].
6.1.3. Lithium
Lithium (Li+) has been shown to significantly decrease cognitive decline in patients with AD without showing any significant adverse effects [169]. In another study of participants with amnestic mild cognitive impairment, long-term lithium treatment reduced p-tau concentration in cerebrospinal fluid and improved performance on the cognitive subscale of the Alzheimer’s Disease Assessment Scale [170]. Recent studies address the effects of lithium leading to cognitive enhancements may due to the induction of autophagy [171, 172]. By inhibitingGSK-3β activity, lithium triggered an autophagic response and modified biological cascades pertaining to the pathophysiology of AD.
6.1.4. Memantine
Memantine is a non-competitive NMDA receptor antagonist, which has been approved for the treatment of moderate-to-severe AD [173]. Recently, from a screening of 796 chemicals, memantine was identified as a novel autophagy inducer with better tolerance and much safer effects. The group found memantine to enhance clearance of damaged mitochondria via the upregulation of autophagy in SH-SY5Y cells illuminating the possible neuroprotective mechanism of memantine [174]. Memantine also demonstrated neuroprotective effects by inducing autophagy via inhibition of the survival PI3K/AKT/mTOR pathway in an in vitro study [175].
6.1.5. Melatonin
Melatonin (N-acetyl-5-methoxytryptamine) is a neurohormone and secreted principally by the pineal gland. Production of melatonin declines during aging, with levels extremely decreased (to below 5%) in AD [176, 177]. Based on these observations, melatonin has been proposed as a potential biomarker for AD. Earlier investigations identified SIRT1 as a critical downstream effector of melatonin in promoting cognitive functions from autophagy [178, 179]. In a recent trial, melatonin supplementation (using much higher concentrations than physiological concentrations) prevented cognitive decline, decreased Tau hyperphosphorylation, and recovered the impaired autophagy flux in the rat and human Tauopathy models [180].
6.1.6. Metformin
Metformin is the first-line oral anti-diabetic drug for type II diabetes. By disturbing complex I of the electron transport chain, metformin induces expression of AMPK and SIRT1, which is thought to be the related to its mechanism of action [181, 182]. The AMPK–mTOR and SIRT1 pathways are the traditional regulatory pathways for autophagy. Metformin also decreases the inhibitory interaction with cytosolic p53 by facilitating Parkin-mediated mitophagy [183]. Consistently, metformin may be a novel activator of autophagy. Recently, a clinical trial showed that long-term treatment of metformin in type II diabetes significantly lowered the incidence of dementia [184, 185]. Chen et al., [186]. further claimed that metformin reduced Tauopathy and improved cognitive impairment by activating AMPK-dependent autophagy. Although a protective effect of metformin in AD has been shown in vivo and in vitro, several studies have demonstrated the opposite viewpoint, and shown that metformin may exacerbate AD progression [187, 188]. Thus, there are still confounding issues at present before a definitive relationship between metformin and AD can be established. Further investigation is required to solidify or refute this relationship
6.1.7. Vitamin D and VDR
Epidemiological data show that serum vitamin D concentrations correlated negatively with the severity of AD. Further, vitamin D deficiency is surfaced to be a risk factor for cognitive decline [189]. Vitamin D modulates its biological effects by hydroxylation, resulting in its biologically active form, 1,25-dyhydroxyvitamin D3(1,25(OH)2D3), which signals through its receptor (VDR) [190]. It has been reported that 1,25(OH)2D3 increased Aβ1–40 vectorial transport across the blood-brain barrier, which reinforced the systemic clearance of Aβ1–40 [39, 191]. Additionally, long-term treatment of mice with 1,25(OH)2D3 during a period of plaque formation reduced soluble and insoluble Aβ, particularly in the hippocampus, in which VDR are abundant. This additionally led to improvements in conditioned fear memory [192]. Recently, vitamin D was reported as a potent inducer of autophagy and indeed may suppress Aβ-induced toxicity in this way. Mechanistically, VDR constitutively represses autophagy. Specifically, upon 1,25(OH)2D3 stimulation, basal levels of autophagy increased by the de-repression of the key autophagy genes, namely, LC3-II and Beclin-1 [193]. The mechanism of 1,25(OH)2D3-induced autophagy also involved AMPK pathway which was triggered by calcium/calmodulin-dependent protein kinase kinase 2 (CAMKK2) activation [194].
6.1.8. Trehalose
Trehalose is a natural disaccharide derived by condensation of two glucose molecules. The beneficial effects of trehalose are related to autophagy induction in an mTOR-independent pathway [195]. In APP/PS1 transgenic mice, trehalose interferes with the formation of Aβ peptide, which significantly reduced the number of amyloid deposits and inhibited the neurotoxicity of Aβ [196]. Besides the selective targeting to Aβ peptides, long-term treatment of trehalose also decreased Tau inclusions in neurons, with a concomitant reduction in p62 levels, suggesting the relevance of autophagy in modulating Tau pathology [197]. Given that trehalose is a safe food ingredient for human use and is Food and Drug Administration “GRAS” (Generally Recognized As Safe), we believe trehalose is a promising agent for further pharmacological investigation in patients with AD.
6.2. Natural Extracts
6.2.1. Resveratrol
Resveratrol(trans-3,4′,5-trihydroxystilbene) displays neuroprotective effects by enhancing autophagy via controlling SIRT1-mediated transcriptional regulation in vivo [118, 119, 179]. Resveratrol also mediates AMPK-dependent or mTOR-dependent autophagy in mice, and thus is associated with anti-amyloidogenicity [198, 199]. In Tau transgenic mice, resveratrol exhibited a strong tendency to decrease tangle deposition along with the reduction of Tau hyperphosphorylation [200]. Although resveratrol is very safe, there are problems related to its clinical use. One is its short half-life in serum, with another being its low oral bioavailability. Recently, long-term use with adequate dosages appears to be an effective way of improving its bioavailability. Nonetheless, further work is still needed to find out how to prolong the short half-life of resveratrol.
6.2.2. Quercetin
Quercetin (3,3′,4′,5,7-pentahydroxyflavone) has been shown to promote learning and memory performance [201] and inhibit the formation of Aβ peptides by regulating BACE-1 activity in AD rats [202]. A recent report showed that the anti-amyloidogenic effect of quercetin is associated with autophagy activation [203]. The NRF2 pathway is the major signaling component in mediating the neuroprotective effect of quercetin. Li et al., demonstrated that quercetin improved cognitive memory by reducing Aβ1-42 levels and increasing expression of NRF2/ heme oxygenase-1 in rat brain [204]. For tau pathology, quercetin reduced hyperphosphorylation of Tau protein by inhibiting GSK3β activity [205]. Consistently, quercetin may be a novel therapeutic agent for AD. As a caveat, low efficiency through the blood-brain barrier and easy metabolism have limited its clinical application. To overcome these limitations, a quercetin modified polysorbate 80-coated gold and palladium core-shell structured nanoparticle has been synthesized. This quercetin modified nanoparticle activated autophagy, accelerated Aβ clearance, and diminished Aβ-mediated cytotoxicity more efficiently than quercetin alone [206].
6.2.3. Ginsenosides
In AD mice, ginsenoside Rb1 improved learning ability and reduced Aβ1-42 in the cortex and hippocampus. Using laser scanning confocal microscopy, Guo et al., [207]. observed LC3 dispersed throughout the cell and at the membrane of autophagosomes after ginsenoside administration. This indicates that autophagy activation is involved in the Aβ-scavenging effects of ginsenosides. mTOR and NRF2 are recognized as the dominant signaling pathways mediating the neuroprotective role of ginsenosides in attenuating Aβ formation [207, 208]. A study further demonstrated that mTOR-dependent TFEB‐mediated autophagy maybe another key pathway facilitating the anti-amyloidogenic effect of ginsenoside K [209]. Ginsenoside Rg1 administration also reduced tau accumulation in a rat model by decreasing GSK3β pathway activation [210].
6.2.4. Curcumin
Curcumin (bis‐α, β‐unsaturated β‐diketone) is a natural compound traditionally used for its anti-tumorigenic and anti-hyperlipidemic properties. The therapeutic function of curcumin in promoting Aβ clearance and ameliorating symptoms of AD in vivo and in vitro has received wide attention [211, 212]. In APP/PS1 transgenic mice, curcumin reduced Aβ generation, induced autophagy, and upregulated expression of Beclin-1 and LC3 I/II [213]. The neuroprotective effect of curcumin is associated with inhibition of the PI3K/AKT/mTOR pathway [213]. Even although the therapeutic function of curcumin has been demonstrated, its poor absorption and low bioavailability have curtailed its clinical application [214]. Compound C1, a monocarbonyl analog of curcumin with a stable structure and good blood–brain barrier permeability, can potently induce nuclear translocation of endogenous TFEB and enhance autophagy in Neuro-2a cells, without inhibiting mTOR activity. Consequently, this may be a novel drug candidate for AD [215].
6.3. Lifestyle Interventions
6.3.1. Calorie Restriction and Fasting
Calorie restriction (CR) (consuming calories 10%–40% less than an ad libitum animal, yet maintaining proper nutrient intake) is generally known as one of the best interventions for slowing aging and increasing lifespan [216]. However, as CR is difficult to perform in practice and easy to cause malnutrition, intermittent fasting (IF) is considered an optimal intervention [217, 218]. Both CR and IF interventions can ameliorate behavioral deficits in 3×TgAD mice. Although only CR affected Aβ1–42 and tau levels in the hippocampus, IF also showed neuroprotective effects against excitotoxic insults and promoted synaptic plasticity [219]. Because inhibition of autophagy attenuates the anti-aging effect of CR and IF, it has been hypothesized that autophagy plays a central role in CR/IF-mediated longevity [220]. In 5×FAD mice, time-lapse live imaging showed a marked increase in levels of EGFP-LC3 vesicles after fasting treatment, which implies that autophagy is activated under starvation. Moreover, starvation-induced autophagy also reduced extracellular Aβ and increased intracellular Aβ degradation [221]. Further, it has been shown that CR ameliorated memory deficits and prevented impairment of adult neurogenesis by restoring autophagic flux in transgenic mice [222]. The IGF-1 and SIRT1 pathways are two widely accepted theories to explain the mechanisms underlying the protective effects of CR. Upon CR intervention, mRNA levels of IGF-1 in the hippocampus are elevated [223]. Similarly, CR also promoted the non-amyloidogenic pathway of APP and attenuated Aβ generation by reducing SIRT1 expression in Tg2576 mice, another mouse model of AD [115, 224].
6.3.2. Physical Activity
Regular physical activity can enhance brain vitality and provides cognitive and psychological benefits [225]. A large prospective study found that exercise improved cognitive and physical performance in patients with AD, with possible additional benefits of reducing neuropsychiatric symptoms [226]. Furthermore, APOE4 allele carriers benefited from exercise intervention on cognitive tasks [227]. Many studies suggest the therapeutic role of physical activity is mediated through autophagy. Swimming training improved learning and memory impairments by downregulating autophagy-related proteins including LC3, Beclin-1, and ATG7, and upregulating p62 in aging rats [228]. Late running reduced Aβ plaque burden with upregulation of autophagy markers (Beclin-1 and syntaxin 17) in the neocortex and hippocampus of AD mice [229]. Regular aerobic exercise delayed the progress of aging and upregulated striatal autophagy via the AMPK/SIRT2 pathway [230]. Although exercise benefited neurocognition in aging models, it is important to note that the duration or intensity of exercise did not matter for the beneficial effects [231]. The best exercise for elderly is in accordance with their cardiorespiratory function.
Conclusion and Perspective
In this review, we have summarized the key role of autophagy in the pathogenesis of AD and discussed potential therapeutic strategies to delay cognitive decline by restoring autophagy. However, applying this strategy in clinical treatment is still some time away. First, long-term or overly activated autophagy may be harmful for cells and may result in cell death. A compound that efficiently activates autophagy but is also safe with long-term use is highly desirable. Second, obstacles of a pharmacological, technical or experimental nature have hampered the straightforward implementation of autophagy modulators in the clinic. One major challenge is that several pro-autophagic drugs not only independently activate autophagy but also potentially disturb other cellular functions, resulting in adverse effects. Third, it is difficult to monitor autophagic flux feasibility and assess therapeutic efficiency of autophagy modulation. To date, immunohistochemical staining against autophagy-specific biomarkers and transmission electron microscopy is a frequently used method to monitor autophagy. However, neither can interpret the results in vivo in a real-time manner. To be applied in a clinical setting, a biomarker or an autophagy-specific surrogate is needed to efficiently assess autophagy modulation. Fourth, how to precisely target the autophagic machinery in the correct tissue and avoid adverse effects associated with systemic autophagy is another big challenge. Nanomedicine maybe a feasible approach, delivering drugs to the targeted sites with a low dose. Nanocarriers can also penetrate through the blood-brain barrier thereby improving bioavailability of the drugs in the brain tissue. Thus, although tremendous advances have been made in our understanding of autophagy, many unanswered questions remain. Much work is needed before we can hope to harness the full therapeutic potential of autophagy modulators.
Acknowledgements
Declared none.
Consent for Publication
Not applicable.
Funding
None.
Conflict of Interest
The authors declare no conflict of interest, financial or otherwise.
References
- 1.Blennow K., de Leon M.J., Zetterberg H. Alzheimer’s disease. Lancet. 2006;368(9533):387–403. doi: 10.1016/S0140-6736(06)69113-7. [DOI] [PubMed] [Google Scholar]
- 2.Bastin M.E., Muñoz M.S., Ferguson K.J., Brown L.J., Wardlaw J.M., MacLullich A.M., Clayden J.D. Quantifying the effects of normal ageing on white matter structure using unsupervised tract shape modelling. Neuroimage. 2010;51(1):1–10. doi: 10.1016/j.neuroimage.2010.02.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Vassar R., Kovacs D.M., Yan R., Wong P.C. The beta-secretase enzyme BACE in health and Alzheimer’s disease: regulation, cell biology, function, and therapeutic potential. J. Neurosci. 2009;29(41):12787–12794. doi: 10.1523/JNEUROSCI.3657-09.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.De Matteis M.A., Luini A. Exiting the golgi complex. Nat. Rev. Mol. Cell Biol. 2008;9(4):273–284. doi: 10.1038/nrm2378. [DOI] [PubMed] [Google Scholar]
- 5.Polito V.A., Li H., Martini-Stoica H., Wang B., Yang L., Xu Y., Swartzlander D.B., Palmieri M., di Ronza A., Lee V.M., Sardiello M., Ballabio A., Zheng H. Selective clearance of aberrant tau proteins and rescue of neurotoxicity by transcription factor EB. EMBO Mol. Med. 2014;6(9):1142–1160. doi: 10.15252/emmm.201303671. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Giannakopoulos P., Herrmann F.R., Bussière T., Bouras C., Kövari E., Perl D.P., Morrison J.H., Gold G., Hof P.R. Tangle and neuron numbers, but not amyloid load, predict cognitive status in Alzheimer’s disease. Neurology. 2003;60(9):1495–1500. doi: 10.1212/01.WNL.0000063311.58879.01. [DOI] [PubMed] [Google Scholar]
- 7.Meyer-Luehmann M., Coomaraswamy J., Bolmont T., Kaeser S., Schaefer C., Kilger E., Neuenschwander A., Abramowski D., Frey P., Jaton A.L., Vigouret J.M., Paganetti P., Walsh D.M., Mathews P.M., Ghiso J., Staufenbiel M., Walker L.C., Jucker M. Exogenous induction of cerebral beta-amyloidogenesis is governed by agent and host. Science. 2006;313(5794):1781–1784. doi: 10.1126/science.1131864. [DOI] [PubMed] [Google Scholar]
- 8.Clavaguera F., Bolmont T., Crowther R.A., Abramowski D., Frank S., Probst A., Fraser G., Stalder A.K., Beibel M., Staufenbiel M., Jucker M., Goedert M., Tolnay M. Transmission and spreading of tauopathy in transgenic mouse brain. Nat. Cell Biol. 2009;11(7):909–913. doi: 10.1038/ncb1901. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Braak H., Braak E. Neuropathological stageing of Alzheimer-related changes. Acta Neuropathol. 1991;82(4):239–259. doi: 10.1007/BF00308809. [DOI] [PubMed] [Google Scholar]
- 10.Papandreou M.E., Tavernarakis N. Autophagy and the endo/exosomal pathways in health and disease. Biotechnol. J. 2017;12(1) doi: 10.1002/biot.201600175. [DOI] [PubMed] [Google Scholar]
- 11.Miranda A.M., Lasiecka Z.M., Xu Y., Neufeld J., Shahriar S., Simoes S., Chan R.B., Oliveira T.G., Small S.A., Di Paolo G. Neuronal lysosomal dysfunction releases exosomes harboring APP C-terminal fragments and unique lipid signatures. Nat. Commun. 2018;9(1):291. doi: 10.1038/s41467-017-02533-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Goetzl E.J., Boxer A., Schwartz J.B., Abner E.L., Petersen R.C., Miller B.L., Kapogiannis D. Altered lysosomal proteins in neural-derived plasma exosomes in preclinical Alzheimer disease. Neurology. 2015;85(1):40–47. doi: 10.1212/WNL.0000000000001702. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Klionsky D.J. Autophagy: from phenomenology to molecular understanding in less than a decade. Nat. Rev. Mol. Cell Biol. 2007;8(11):931–937. doi: 10.1038/nrm2245. [DOI] [PubMed] [Google Scholar]
- 14.Ostertag M., Stammler J., Douchkov D., Eichmann R., Hückelhoven R. The conserved oligomeric Golgi complex is involved in penetration resistance of barley to the barley powdery mildew fungus. Mol. Plant Pathol. 2013;14(3):230–240. doi: 10.1111/j.1364-3703.2012.00846.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Yen W.L., Shintani T., Nair U., Cao Y., Richardson B.C., Li Z., Hughson F.M., Baba M., Klionsky D.J. The conserved oligomeric Golgi complex is involved in double-membrane vesicle formation during autophagy. J. Cell Biol. 2010;188(1):101–114. doi: 10.1083/jcb.200904075. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Eskelinen E.L., Saftig P. Autophagy: a lysosomal degradation pathway with a central role in health and disease. Biochim. Biophys. Acta. 2009;1793(4):664–673. doi: 10.1016/j.bbamcr.2008.07.014. [DOI] [PubMed] [Google Scholar]
- 17.Mizushima N., Levine B., Cuervo A.M., Klionsky D.J. Autophagy fights disease through cellular self-digestion. Nature. 2008;451(7182):1069–1075. doi: 10.1038/nature06639. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Zare-Shahabadi A., Masliah E., Johnson G.V., Rezaei N. Autophagy in Alzheimer’s disease. Rev. Neurosci. 2015;26(4):385–395. doi: 10.1515/revneuro-2014-0076. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Xu J., Camfield R., Gorski S.M. The interplay between exosomes and autophagy - partners in crime. J. Cell Sci. 2018;131(15):jcs215210. doi: 10.1242/jcs.215210. [DOI] [PubMed] [Google Scholar]
- 20.Kuang H., Tan C.Y., Tian H.Z., Liu L.H., Yang M.W., Hong F.F., Yang S.L. Exploring the bi-directional relationship between autophagy and Alzheimer’s disease. CNS Neurosci. Ther. 2019;••• doi: 10.1111/cns.13216. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Menzies F.M., Fleming A., Rubinsztein D.C. Compromised autophagy and neurodegenerative diseases. Nat. Rev. Neurosci. 2015;16(6):345–357. doi: 10.1038/nrn3961. [DOI] [PubMed] [Google Scholar]
- 22.Rubinsztein D.C., Codogno P., Levine B. Autophagy modulation as a potential therapeutic target for diverse diseases. Nat. Rev. Drug Discov. 2012;11(9):709–730. doi: 10.1038/nrd3802. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Korolchuk V.I., Menzies F.M., Rubinsztein D.C. Mechanisms of cross-talk between the ubiquitin-proteasome and autophagy-lysosome systems. FEBS Lett. 2010;584(7):1393–1398. doi: 10.1016/j.febslet.2009.12.047. [DOI] [PubMed] [Google Scholar]
- 24.Agarwal S., Tiwari S.K., Seth B., Yadav A., Singh A., Mudawal A., Chauhan L.K., Gupta S.K., Choubey V., Tripathi A., Kumar A., Ray R.S., Shukla S., Parmar D., Chaturvedi R.K. Activation of autophagic flux against Xenoestrogen Bisphenol-A-induced hippocampal neurodegeneration via AMP kinase (AMPK)/Mammalian target of rapamycin (mTOR) pathways. J. Biol. Chem. 2015;290(34):21163–21184. doi: 10.1074/jbc.M115.648998. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 25.Alers S., Löffler A.S., Wesselborg S., Stork B. Role of AMPK-mTOR-Ulk1/2 in the regulation of autophagy: cross talk, shortcuts, and feedbacks. Mol. Cell. Biol. 2012;32(1):2–11. doi: 10.1128/MCB.06159-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Nixon R.A. The role of autophagy in neurodegenerative disease. Nat. Med. 2013;19(8):983–997. doi: 10.1038/nm.3232. [DOI] [PubMed] [Google Scholar]
- 27.Füllgrabe J., Klionsky D.J., Joseph B. The return of the nucleus: transcriptional and epigenetic control of autophagy. Nat. Rev. Mol. Cell Biol. 2014;15(1):65–74. doi: 10.1038/nrm3716. [DOI] [PubMed] [Google Scholar]
- 28.Obara K., Ohsumi Y. Dynamics and function of PtdIns(3)P in autophagy. Autophagy. 2008;4(7):952–954. doi: 10.4161/auto.6790. [DOI] [PubMed] [Google Scholar]
- 29.Puri C., Renna M., Bento C.F., Moreau K., Rubinsztein D.C. Diverse autophagosome membrane sources coalesce in recycling endosomes. Cell. 2013;154(6):1285–1299. doi: 10.1016/j.cell.2013.08.044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Zhou L., Wang H.F., Ren H.G., Chen D., Gao F., Hu Q.S., Fu C., Xu R.J., Ying Z., Wang G.H. Bcl-2-dependent upregulation of autophagy by sequestosome 1/p62 in vitro. Acta Pharmacol. Sin. 2013;34(5):651–656. doi: 10.1038/aps.2013.12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Lee S., Sato Y., Nixon R.A. Lysosomal proteolysis inhibition selectively disrupts axonal transport of degradative organelles and causes an Alzheimer’s-like axonal dystrophy. J. Neurosci. 2011;31(21):7817–7830. doi: 10.1523/JNEUROSCI.6412-10.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Nilsson P., Loganathan K., Sekiguchi M., Matsuba Y., Hui K., Tsubuki S., Tanaka M., Iwata N., Saito T., Saido T.C. Aβ secretion and plaque formation depend on autophagy. Cell Rep. 2013;5(1):61–69. doi: 10.1016/j.celrep.2013.08.042. [DOI] [PubMed] [Google Scholar]
- 33.Rocchi A., Yamamoto S., Ting T., Fan Y., Sadleir K., Wang Y., Zhang W., Huang S., Levine B., Vassar R., He C.A. Becn1 mutation mediates hyperactive autophagic sequestration of amyloid oligomers and improved cognition in Alzheimer’s disease. PLoS Genet. 2017;13(8):e1006962. doi: 10.1371/journal.pgen.1006962. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Leissring M.A. Aβ-degrading proteases: Therapeutic Potential in Alzheimer disease. CNS Drugs. 2016;30(8):667–675. doi: 10.1007/s40263-016-0364-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Cho M.H., Cho K., Kang H.J., Jeon E.Y., Kim H.S., Kwon H.J., Kim H.M., Kim D.H., Yoon S.Y. Autophagy in microglia degrades extracellular β-amyloid fibrils and regulates the NLRP3 inflammasome. Autophagy. 2014;10(10):1761–1775. doi: 10.4161/auto.29647. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Tian Y., Bustos V., Flajolet M., Greengard P. A small-molecule enhancer of autophagy decreases levels of Abeta and APP-CTF via Atg5-dependent autophagy pathway. FASEB J. 2011;25(6):1934–1942. doi: 10.1096/fj.10-175158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Nilsson P., Saido T.C. Dual roles for autophagy: degradation and secretion of Alzheimer’s disease Aβ peptide. BioEssays. 2014;36(6):570–578. doi: 10.1002/bies.201400002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Pickford F., Masliah E., Britschgi M., Lucin K., Narasimhan R., Jaeger P.A., Small S., Spencer B., Rockenstein E., Levine B., Wyss-Coray T. The autophagy-related protein beclin 1 shows reduced expression in early Alzheimer disease and regulates amyloid beta accumulation in mice. J. Clin. Invest. 2008;118(6):2190–2199. doi: 10.1172/JCI33585. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Guerreiro R.J., Gustafson D.R., Hardy J. The genetic architecture of Alzheimer’s disease: beyond APP, PSENs and APOE. Neurobiol. Aging. 2012;33(3):437–456. doi: 10.1016/j.neurobiolaging.2010.03.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Lee J.H., Yu W.H., Kumar A., Lee S., Mohan P.S., Peterhoff C.M., Wolfe D.M., Martinez-Vicente M., Massey A.C., Sovak G., Uchiyama Y., Westaway D., Cuervo A.M., Nixon R.A. Lysosomal proteolysis and autophagy require presenilin 1 and are disrupted by Alzheimer-related PS1 mutations. Cell. 2010;141(7):1146–1158. doi: 10.1016/j.cell.2010.05.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Belinson H., Lev D., Masliah E., Michaelson D.M. Activation of the amyloid cascade in apolipoprotein E4 transgenic mice induces lysosomal activation and neurodegeneration resulting in marked cognitive deficits. J. Neurosci. 2008;28(18):4690–4701. doi: 10.1523/JNEUROSCI.5633-07.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Ji Z.S., Müllendorff K., Cheng I.H., Miranda R.D., Huang Y., Mahley R.W. Reactivity of apolipoprotein E4 and amyloid beta peptide: lysosomal stability and neurodegeneration. J. Biol. Chem. 2006;281(5):2683–2692. doi: 10.1074/jbc.M506646200. [DOI] [PubMed] [Google Scholar]
- 43.Goedert M., Spillantini M.G. A century of Alzheimer’s disease. Science. 2006;314(5800):777–781. doi: 10.1126/science.1132814. [DOI] [PubMed] [Google Scholar]
- 44.Bednarski E., Lynch G. Cytosolic proteolysis of tau by cathepsin D in hippocampus following suppression of cathepsins B and L. J. Neurochem. 1996;67(5):1846–1855. doi: 10.1046/j.1471-4159.1996.67051846.x. [DOI] [PubMed] [Google Scholar]
- 45.Hamano T., Gendron T.F., Causevic E., Yen S.H., Lin W.L., Isidoro C., Deture M., Ko L.W. 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]
- 46.Lee M.J., Lee J.H., Rubinsztein D.C. Tau degradation: the ubiquitin-proteasome system versus the autophagy-lysosome system. Prog. Neurobiol. 2013;105:49–59. doi: 10.1016/j.pneurobio.2013.03.001. [DOI] [PubMed] [Google Scholar]
- 47.Hanger D.P., Hughes K., Woodgett J.R., Brion J.P., Anderton B.H. Glycogen synthase kinase-3 induces Alzheimer’s disease-like phosphorylation of tau: generation of paired helical filament epitopes and neuronal localisation of the kinase. Neurosci. Lett. 1992;147(1):58–62. doi: 10.1016/0304-3940(92)90774-2. [DOI] [PubMed] [Google Scholar]
- 48.Zhou Y., Hayashi I., Wong J., Tugusheva K., Renger J.J., Zerbinatti C. Intracellular clusterin interacts with brain isoforms of the bridging integrator 1 and with the microtubule-associated protein Tau in Alzheimer’s disease. PLoS One. 2014;9(7):e103187. doi: 10.1371/journal.pone.0103187. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Lee S.J., Desplats P., Sigurdson C., Tsigelny I., Masliah E. Cell-to-cell transmission of non-prion protein aggregates. Nat. Rev. Neurol. 2010;6(12):702–706. doi: 10.1038/nrneurol.2010.145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Sardar Sinha M., Ansell-Schultz A., Civitelli L., Hildesjö C., Larsson M., Lannfelt L., Ingelsson M., Hallbeck M. Alzheimer’s disease pathology propagation by exosomes containing toxic amyloid-beta oligomers. Acta Neuropathol. 2018;136(1):41–56. doi: 10.1007/s00401-018-1868-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Eisele Y.S., Bolmont T., Heikenwalder M., Langer F., Jacobson L.H., Yan Z.X., Roth K., Aguzzi A., Staufenbiel M., Walker L.C., Jucker M. Induction of cerebral beta-amyloidosis: intracerebral versus systemic Abeta inoculation. Proc. Natl. Acad. Sci. USA. 2009;106(31):12926–12931. doi: 10.1073/pnas.0903200106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Asai H., Ikezu S., Tsunoda S., Medalla M., Luebke J., Haydar T., Wolozin B., Butovsky O., Kügler S., Ikezu T. Depletion of microglia and inhibition of exosome synthesis halt tau propagation. Nat. Neurosci. 2015;18(11):1584–1593. doi: 10.1038/nn.4132. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Rajendran L., Honsho M., Zahn T.R., Keller P., Geiger K.D., Verkade P., Simons K. Alzheimer’s disease beta-amyloid peptides are released in association with exosomes. Proc. Natl. Acad. Sci. USA. 2006;103(30):11172–11177. doi: 10.1073/pnas.0603838103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Codogno P., Meijer A.J. Autophagy and signaling: their role in cell survival and cell death. Cell Death Differ. 2005;12(Suppl. 2):1509–1518. doi: 10.1038/sj.cdd.4401751. [DOI] [PubMed] [Google Scholar]
- 55.Ma T., Hoeffer C.A., Capetillo-Zarate E., Yu F., Wong H., Lin M.T., Tampellini D., Klann E., Blitzer R.D., Gouras G.K. Dysregulation of the mTOR pathway mediates impairment of synaptic plasticity in a mouse model of Alzheimer’s disease. PLoS One. 2010;5(9):e12845. doi: 10.1371/journal.pone.0012845. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Paccalin M., Pain-Barc S., Pluchon C., Paul C., Besson M.N., Carret-Rebillat A.S., Rioux-Bilan A., Gil R., Hugon J. Activated mTOR and PKR kinases in lymphocytes correlate with memory and cognitive decline in Alzheimer’s disease. Dement. Geriatr. Cogn. Disord. 2006;22(4):320–326. doi: 10.1159/000095562. [DOI] [PubMed] [Google Scholar]
- 57.Lafay-Chebassier C., Paccalin M., Page G., Barc-Pain S., Perault-Pochat M.C., Gil R., Pradier L., Hugon J. mTOR/ p70S6k signalling alteration by Abeta exposure as well as in APP-PS1 transgenic models and in patients with Alzheimer’s disease. J. Neurochem. 2005;94(1):215–225. doi: 10.1111/j.1471-4159.2005.03187.x. [DOI] [PubMed] [Google Scholar]
- 58.Cai Z., Chen G., He W., Xiao M., Yan L.J. Activation of mTOR: a culprit of Alzheimer’s disease? Neuropsychiatr. Dis. Treat. 2015;11:1015–1030. doi: 10.2147/NDT.S75717. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Caccamo A., Magrì A., Medina D.X., Wisely E.V., López-Aranda M.F., Silva A.J., Oddo S. mTOR regulates tau phosphorylation and degradation: implications for Alzheimer’s disease and other tauopathies. Aging Cell. 2013;12(3):370–380. doi: 10.1111/acel.12057. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Congdon E.E., Wu J.W., Myeku N., Figueroa Y.H., Herman M., Marinec P.S., Gestwicki J.E., Dickey C.A., Yu W.H., Duff K.E. Methylthioninium chloride (methylene blue) induces autophagy and attenuates tauopathy in vitro and in vivo. Autophagy. 2012;8(4):609–622. doi: 10.4161/auto.19048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Spilman P., Podlutskaya N., Hart M.J., Debnath J., Gorostiza O., Bredesen D., Richardson A., Strong R., Galvan V. Inhibition of mTOR by rapamycin abolishes cognitive deficits and reduces amyloid-beta levels in a mouse model of Alzheimer’s disease. PLoS One. 2010;5(4):e9979. doi: 10.1371/journal.pone.0009979. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Maiese K., Chong Z.Z., Shang Y.C., Wang S. mTOR: on target for novel therapeutic strategies in the nervous system. Trends Mol. Med. 2013;19(1):51–60. doi: 10.1016/j.molmed.2012.11.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Cai Z., Yan L.J., Li K., Quazi S.H., Zhao B. Roles of AMP-activated protein kinase in Alzheimer’s disease. Neuromolecular Med. 2012;14(1):1–14. doi: 10.1007/s12017-012-8173-2. [DOI] [PubMed] [Google Scholar]
- 64.O’Neill C., Kiely A.P., Coakley M.F., Manning S., Long-Smith C.M. Insulin and IGF-1 signalling: longevity, protein homoeostasis and Alzheimer’s disease. Biochem. Soc. Trans. 2012;40(4):721–727. doi: 10.1042/BST20120080. [DOI] [PubMed] [Google Scholar]
- 65.Dibble C.C., Cantley L.C. Regulation of mTORC1 by PI3K signaling. Trends Cell Biol. 2015;25(9):545–555. doi: 10.1016/j.tcb.2015.06.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Mabuchi S., Kuroda H., Takahashi R., Sasano T. The PI3K/AKT/mTOR pathway as a therapeutic target in ovarian cancer. Gynecol. Oncol. 2015;137(1):173–179. doi: 10.1016/j.ygyno.2015.02.003. [DOI] [PubMed] [Google Scholar]
- 67.Tanida I. Autophagosome formation and molecular mechanism of autophagy. Antioxid. Redox Signal. 2011;14(11):2201–2214. doi: 10.1089/ars.2010.3482. [DOI] [PubMed] [Google Scholar]
- 68.Perluigi M., Pupo G., Tramutola A., Cini C., Coccia R., Barone E., Head E., Butterfield D.A., Di Domenico F. Neuropathological role of PI3K/Akt/mTOR axis in Down syndrome brain. Biochim. Biophys. Acta. 2014;1842(7):1144–1153. doi: 10.1016/j.bbadis.2014.04.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Griffin R.J., Moloney A., Kelliher M., Johnston J.A., Ravid R., Dockery P., O’Connor R., O’Neill C. Activation of Akt/PKB, increased phosphorylation of Akt substrates and loss and altered distribution of Akt and PTEN are features of Alzheimer’s disease pathology. J. Neurochem. 2005;93(1):105–117. doi: 10.1111/j.1471-4159.2004.02949.x. [DOI] [PubMed] [Google Scholar]
- 70.Rickle A., Bogdanovic N., Volkmann I., Zhou X., Pei J.J., Winblad B., Cowburn R.F. PTEN levels in Alzheimer’s disease medial temporal cortex. Neurochem. Int. 2006;48(2):114–123. doi: 10.1016/j.neuint.2005.08.014. [DOI] [PubMed] [Google Scholar]
- 71.Sonoda Y., Mukai H., Matsuo K., Takahashi M., Ono Y., Maeda K., Akiyama H., Kawamata T. Accumulation of tumor-suppressor PTEN in Alzheimer neurofibrillary tangles. Neurosci. Lett. 2010;471(1):20–24. doi: 10.1016/j.neulet.2009.12.078. [DOI] [PubMed] [Google Scholar]
- 72.Wani A., Gupta M., Ahmad M., Shah A.M., Ahsan A.U., Qazi P.H., Malik F., Singh G., Sharma P.R., Kaddoumi A., Bharate S.B., Vishwakarma R.A., Kumar A. Alborixin clears amyloid-β by inducing autophagy through PTEN-mediated inhibition of the AKT pathway. Autophagy. 2019;15(10):1810–1828. doi: 10.1080/15548627.2019.1596476. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Zhang X., Li F., Bulloj A., Zhang Y.W., Tong G., Zhang Z., Liao F.F., Xu H. Tumor-suppressor PTEN affects tau phosphorylation, aggregation, and binding to microtubules. FASEB J. 2006;20(8):1272–1274. doi: 10.1096/fj.06-5721fje. [DOI] [PubMed] [Google Scholar]
- 74.Stretton C., Hoffmann T.M., Munson M.J., Prescott A., Taylor P.M., Ganley I.G., Hundal H.S. GSK3-mediated raptor phosphorylation supports amino-acid-dependent mTORC1-directed signalling. Biochem. J. 2015;470(2):207–221. doi: 10.1042/BJ20150404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Kirouac L., Rajic A.J., Cribbs D.H., Padmanabhan J. Activation of Ras-ERK signaling and GSK-3 by amyloid precursor protein and amyloid beta facilitates neurodegeneration in Alzheimer’s Disease. 2017. [DOI] [PMC free article] [PubMed]
- 76.Terwel D., Muyllaert D., Dewachter I., Borghgraef P., Croes S., Devijver H., Van Leuven F. Amyloid activates GSK-3beta to aggravate neuronal tauopathy in bigenic mice. Am. J. Pathol. 2008;172(3):786–798. doi: 10.2353/ajpath.2008.070904. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Shi X.L., Wu J.D., Liu P., Liu Z.P. Synthesis and evaluation of novel GSK-3β inhibitors as multifunctional agents against Alzheimer’s disease. Eur. J. Med. Chem. 2019;167:211–225. doi: 10.1016/j.ejmech.2019.02.001. [DOI] [PubMed] [Google Scholar]
- 78.Gavilán E., Pintado C., Gavilan M.P., Daza P., Sánchez-Aguayo I., Castaño A., Ruano D. Age-related dysfunctions of the autophagy lysosomal pathway in hippocampal pyramidal neurons under proteasome stress. Neurobiol. Aging. 2015;36(5):1953–1963. doi: 10.1016/j.neurobiolaging.2015.02.025. [DOI] [PubMed] [Google Scholar]
- 79.Kim J., Kundu M., Viollet B., Guan K.L. AMPK and mTOR regulate autophagy through direct phosphorylation of Ulk1. Nat. Cell Biol. 2011;13(2):132–141. doi: 10.1038/ncb2152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Shang L., Chen S., Du F., Li S., Zhao L., Wang X. Nutrient starvation elicits an acute autophagic response mediated by Ulk1 dephosphorylation and its subsequent dissociation from AMPK. Proc. Natl. Acad. Sci. USA. 2011;108(12):4788–4793. doi: 10.1073/pnas.1100844108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Danielpour D., Gao Z., Zmina P.M., Shankar E., Shultes B.C., Jobava R., Welford S.M., Hatzoglou M. Early cellular responses of prostate carcinoma cells to sepantronium bromide (YM155) Involve suppression of mTORC1 by AMPK. Sci. Rep. 2019;9(1):11541. doi: 10.1038/s41598-019-47573-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Löffler A.S., Alers S., Dieterle A.M., Keppeler H., Franz-Wachtel M., Kundu M., Campbell D.G., Wesselborg S., Alessi D.R., Stork B. Ulk1-mediated phosphorylation of AMPK constitutes a negative regulatory feedback loop. Autophagy. 2011;7(7):696–706. doi: 10.4161/auto.7.7.15451. [DOI] [PubMed] [Google Scholar]
- 83.Vingtdeux V., Chandakkar P., Zhao H., d’Abramo C., Davies P., Marambaud P. Novel synthetic small-molecule activators of AMPK as enhancers of autophagy and amyloid-β peptide degradation. FASEB J. 2011;25(1):219–231. doi: 10.1096/fj.10-167361. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Vingtdeux V., Davies P., Dickson D.W., Marambaud P. AMPK is abnormally activated in tangle- and pre-tangle-bearing neurons in Alzheimer’s disease and other tauopathies. Acta Neuropathol. 2011;121(3):337–349. doi: 10.1007/s00401-010-0759-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Avgerinos K.I., Kalaitzidis G., Malli A., Kalaitzoglou D., Myserlis P.G., Lioutas V.A. Intranasal insulin in Alzheimer’s dementia or mild cognitive impairment: a systematic review. J. Neurol. 2018;265(7):1497–1510. doi: 10.1007/s00415-018-8768-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Bains M., Florez-McClure M.L., Heidenreich K.A. Insulin-like growth factor-I prevents the accumulation of autophagic vesicles and cell death in Purkinje neurons by increasing the rate of autophagosome-to-lysosome fusion and degradation. J. Biol. Chem. 2009;284(30):20398–20407. doi: 10.1074/jbc.M109.011791. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Liu Q., Guan J.Z., Sun Y., Le Z., Zhang P., Yu D., Liu Y. Insulin-like growth factor 1 receptor-mediated cell survival in hypoxia depends on the promotion of autophagy via suppression of the PI3K/Akt/mTOR signaling pathway. Mol. Med. Rep. 2017;15(4):2136–2142. doi: 10.3892/mmr.2017.6265. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Kim B., Elzinga S.E., Henn R.E., McGinley L.M., Feldman E.L. The effects of insulin and insulin-like growth factor I on amyloid precursor protein phosphorylation in in vitro and in vivo models of Alzheimer’s disease. Neurobiol. Dis. 2019;132:104541. doi: 10.1016/j.nbd.2019.104541. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Carro E., Trejo J.L., Gomez-Isla T., LeRoith D., Torres-Aleman I. Serum insulin-like growth factor I regulates brain amyloid-beta levels. Nat. Med. 2002;8(12):1390–1397. doi: 10.1038/nm1202-793. [DOI] [PubMed] [Google Scholar]
- 90.Lesort M., Jope R.S., Johnson G.V. Insulin transiently increases tau phosphorylation: involvement of glycogen synthase kinase-3beta and Fyn tyrosine kinase. J. Neurochem. 1999;72(2):576–584. doi: 10.1046/j.1471-4159.1999.0720576.x. [DOI] [PubMed] [Google Scholar]
- 91.Settembre C., Di Malta C., Polito V.A., Garcia Arencibia M., Vetrini F., Erdin S., Erdin S.U., Huynh T., Medina D., Colella P., Sardiello M., Rubinsztein D.C., Ballabio A. TFEB links autophagy to lysosomal biogenesis. Science. 2011;332(6036):1429–1433. doi: 10.1126/science.1204592. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Roczniak-Ferguson A., Petit C.S., Froehlich F., Qian S., Ky J., Angarola B., Walther T.C., Ferguson S.M. The transcription factor TFEB links mTORC1 signaling to transcriptional control of lysosome homeostasis. Sci. Signal. 2012;5(228):ra42. doi: 10.1126/scisignal.2002790. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Settembre C., Zoncu R., Medina D.L., Vetrini F., Erdin S., Erdin S., Huynh T., Ferron M., Karsenty G., Vellard M.C., Facchinetti V., Sabatini D.M., Ballabio A. A lysosome-to-nucleus signalling mechanism senses and regulates the lysosome via mTOR and TFEB. EMBO J. 2012;31(5):1095–1108. doi: 10.1038/emboj.2012.32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Sardiello M., Palmieri M., di Ronza A., Medina D.L., Valenza M., Gennarino V.A., Di Malta C., Donaudy F., Embrione V., Polishchuk R.S., Banfi S., Parenti G., Cattaneo E., Ballabio A. A gene network regulating lysosomal biogenesis and function. Science. 2009;325(5939):473–477. doi: 10.1126/science.1174447. [DOI] [PubMed] [Google Scholar]
- 95.Medina D.L., Di Paola S., Peluso I., Armani A., De Stefani D., Venditti R., Montefusco S., Scotto-Rosato A., Prezioso C., Forrester A., Settembre C., Wang W., Gao Q., Xu H., Sandri M., Rizzuto R., De Matteis M.A., Ballabio A. Lysosomal calcium signalling regulates autophagy through calcineurin and TFEB. Nat. Cell Biol. 2015;17(3):288–299. doi: 10.1038/ncb3114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Xiao Q., Yan P., Ma X., Liu H., Perez R., Zhu A., Gonzales E., Burchett J.M., Schuler D.R., Cirrito J.R., Diwan A., Lee J.M. Enhancing astrocytic lysosome biogenesis facilitates Aβ clearance and attenuates amyloid plaque pathogenesis. J. Neurosci. 2014;34(29):9607–9620. doi: 10.1523/JNEUROSCI.3788-13.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Zhang Y.D., Zhao J.J. TFEB Participates in the Aβ-Induced Pathogenesis of Alzheimer’s Disease by Regulating the Autophagy-Lysosome Pathway. DNA Cell Biol. 2015;34(11):661–668. doi: 10.1089/dna.2014.2738. [DOI] [PubMed] [Google Scholar]
- 98.Chong C.M., Ke M., Tan Y., Huang Z., Zhang K., Ai N., Ge W., Qin D., Lu J.H., Su H. Presenilin 1 deficiency suppresses autophagy in human neural stem cells through reducing γ-secretase-independent ERK/CREB signaling. Cell Death Dis. 2018;9(9):879. doi: 10.1038/s41419-018-0945-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Ohashi Y., Soler N., García Ortegón M., Zhang L., Kirsten M.L., Perisic O., Masson G.R., Burke J.E., Jakobi A.J., Apostolakis A.A., Johnson C.M., Ohashi M., Ktistakis N.T., Sachse C., Williams R.L. Characterization of Atg38 and NRBF2, a fifth subunit of the autophagic Vps34/PIK3C3 complex. Autophagy. 2016;12(11):2129–2144. doi: 10.1080/15548627.2016.1226736. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Ma X., Zhang S., He L., Rong Y., Brier L.W., Sun Q., Liu R., Fan W., Chen S., Yue Z., Kim J., Guan K.L., Li D., Zhong Q. MTORC1-mediated NRBF2 phosphorylation functions as a switch for the class III PtdIns3K and autophagy. Autophagy. 2017;13(3):592–607. doi: 10.1080/15548627.2016.1269988. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Yang C., Cai C.Z., Song J.X., Tan J.Q., Durairajan S.S.K., Iyaswamy A., Wu M.Y., Chen L.L., Yue Z., Li M., Lu J.H. NRBF2 is involved in the autophagic degradation process of APP-CTFs in Alzheimer disease models. Autophagy. 2017;13(12):2028–2040. doi: 10.1080/15548627.2017.1379633. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Saitoh Y., Fujikake N., Okamoto Y., Popiel H.A., Hatanaka Y., Ueyama M., Suzuki M., Gaumer S., Murata M., Wada K., Nagai Y. p62 plays a protective role in the autophagic degradation of polyglutamine protein oligomers in polyglutamine disease model flies. J. Biol. Chem. 2015;290(3):1442–1453. doi: 10.1074/jbc.M114.590281. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Perez S.E., He B., Nadeem M., Wuu J., Ginsberg S.D., Ikonomovic M.D., Mufson E.J. Hippocampal endosomal, lysosomal, and autophagic dysregulation in mild cognitive impairment: correlation with aβ and tau pathology. J. Neuropathol. Exp. Neurol. 2015;74(4):345–358. doi: 10.1097/NEN.0000000000000179. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Seibenhener M.L., Babu J.R., Geetha T., Wong H.C., Krishna N.R., Wooten M.W. Sequestosome 1/p62 is a polyubiquitin chain binding protein involved in ubiquitin proteasome degradation. Mol. Cell. Biol. 2004;24(18):8055–8068. doi: 10.1128/MCB.24.18.8055-8068.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Babu J.R., Geetha T., Wooten M.W. Sequestosome 1/p62 shuttles polyubiquitinated tau for proteasomal degradation. J. Neurochem. 2005;94(1):192–203. doi: 10.1111/j.1471-4159.2005.03181.x. [DOI] [PubMed] [Google Scholar]
- 106.Song P., Li S., Wu H., Gao R., Rao G., Wang D., Chen Z., Ma B., Wang H., Sui N., Deng H., Zhang Z., Tang T., Tan Z., Han Z., Lu T., Zhu Y., Chen Q. Parkin promotes proteasomal degradation of p62: implication of selective vulnerability of neuronal cells in the pathogenesis of Parkinson’s disease. Protein Cell. 2016;7(2):114–129. doi: 10.1007/s13238-015-0230-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Du Y., Wooten M.C., Gearing M., Wooten M.W. Age-associated oxidative damage to the p62 promoter: implications for Alzheimer disease. Free Radic. Biol. Med. 2009;46(4):492–501. doi: 10.1016/j.freeradbiomed.2008.11.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Caccamo A., Ferreira E., Branca C., Oddo S. p62 improves AD-like pathology by increasing autophagy. Mol. Psychiatry. 2017;22(6):865–873. doi: 10.1038/mp.2016.139. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 109.Ramesh Babu J., Lamar Seibenhener M., Peng J., Strom A.L., Kemppainen R., Cox N., Zhu H., Wooten M.C., Diaz-Meco M.T., Moscat J., Wooten M.W. Genetic inactivation of p62 leads to accumulation of hyperphosphorylated tau and neurodegeneration. J. Neurochem. 2008;106(1):107–120. doi: 10.1111/j.1471-4159.2008.05340.x. [DOI] [PubMed] [Google Scholar]
- 110.Wu C.L., Chen C.H., Hwang C.S., Chen S.D., Hwang W.C., Yang D.I. Roles of p62 in BDNF-dependent autophagy suppression and neuroprotection against mitochondrial dysfunction in rat cortical neurons. J. Neurochem. 2017;140(6):845–861. doi: 10.1111/jnc.13937. [DOI] [PubMed] [Google Scholar]
- 111.Johnson R., Shabalala S., Louw J., Kappo A.P., Muller C.J.F. Aspalathin reverts doxorubicin-induced cardiotoxicity through increased autophagy and decreased expression of p53/mTOR/p62 Signaling. Molecules. 2017;22(10):E1589. doi: 10.3390/molecules22101589. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Duran A., Amanchy R., Linares J.F., Joshi J., Abu-Baker S., Porollo A., Hansen M., Moscat J., Diaz-Meco M.T. p62 is a key regulator of nutrient sensing in the mTORC1 pathway. Mol. Cell. 2011;44(1):134–146. doi: 10.1016/j.molcel.2011.06.038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Nihira K., Miki Y., Ono K., Suzuki T., Sasano H. An inhibition of p62/SQSTM1 caused autophagic cell death of several human carcinoma cells. Cancer Sci. 2014;105(5):568–575. doi: 10.1111/cas.12396. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Price N.L., Gomes A.P., Ling A.J., Duarte F.V., Martin-Montalvo A., North B.J., Agarwal B., Ye L., Ramadori G., Teodoro J.S., Hubbard B.P., Varela A.T., Davis J.G., Varamini B., Hafner A., Moaddel R., Rolo A.P., Coppari R., Palmeira C.M., de Cabo R., Baur J.A., Sinclair D.A. SIRT1 is required for AMPK activation and the beneficial effects of resveratrol on mitochondrial function. Cell Metab. 2012;15(5):675–690. doi: 10.1016/j.cmet.2012.04.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Bonda D.J., Lee H.G., Camins A., Pallàs M., Casadesus G., Smith M.A., Zhu X. The sirtuin pathway in ageing and Alzheimer disease: mechanistic and therapeutic considerations. Lancet Neurol. 2011;10(3):275–279. doi: 10.1016/S1474-4422(11)70013-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Lee I.H., Cao L., Mostoslavsky R., Lombard D.B., Liu J., Bruns N.E., Tsokos M., Alt F.W., Finkel T. A role for the NAD-dependent deacetylase Sirt1 in the regulation of autophagy. Proc. Natl. Acad. Sci. USA. 2008;105(9):3374–3379. doi: 10.1073/pnas.0712145105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Luo G., Jian Z., Zhu Y., Zhu Y., Chen B., Ma R., Tang F., Xiao Y. Sirt1 promotes autophagy and inhibits apoptosis to protect cardiomyocytes from hypoxic stress. Int. J. Mol. Med. 2019;43(5):2033–2043. doi: 10.3892/ijmm.2019.4125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Morselli E., Maiuri M.C., Markaki M., Megalou E., Pasparaki A., Palikaras K., Criollo A., Galluzzi L., Malik S.A., Vitale I., Michaud M., Madeo F., Tavernarakis N., Kroemer G. The life span-prolonging effect of sirtuin-1 is mediated by autophagy. Autophagy. 2010;6(1):186–188. doi: 10.4161/auto.6.1.10817. [DOI] [PubMed] [Google Scholar]
- 119.Ge J.F., Qiao J.P., Qi C.C., Wang C.W., Zhou J.N. The binding of resveratrol to monomer and fibril amyloid beta. Neurochem. Int. 2012;61(7):1192–1201. doi: 10.1016/j.neuint.2012.08.012. [DOI] [PubMed] [Google Scholar]
- 120.Min S.W., Sohn P.D., Li Y., Devidze N., Zhou J.N. Johnson, J.R.; Krogan, N.J.; Masliah, E.; Mok, S.A.; Gestwicki, J.E.; Gan, L. SIRT1 deacetylates Tau and reduces pathogenic tau spread in a mouse model of tauopathy. J. Neurosci. 2018;38(15):3680–3688. doi: 10.1523/JNEUROSCI.2369-17.2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Cho S.H., Chen J.A., Sayed F., Ward M.E., Gao F., Nguyen T.A., Krabbe G., Sohn P.D., Lo I., Minami S., Devidze N., Zhou Y., Coppola G., Gan L. SIRT1 deficiency in microglia contributes to cognitive decline in aging and neurodegeneration via epigenetic regulation of IL-1β. J. Neurosci. 2015;35(2):807–818. doi: 10.1523/JNEUROSCI.2939-14.2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Tasdemir E., Maiuri M.C., Galluzzi L., Vitale I., Djavaheri-Mergny M., D’Amelio M., Criollo A., Morselli E., Zhu C., Harper F., Nannmark U., Samara C., Pinton P., Vicencio J.M., Carnuccio R., Moll U.M., Madeo F., Paterlini-Brechot P., Rizzuto R., Szabadkai G., Pierron G., Blomgren K., Tavernarakis N., Codogno P., Cecconi F., Kroemer G. Regulation of autophagy by cytoplasmic p53. Nat. Cell Biol. 2008;10(6):676–687. doi: 10.1038/ncb1730. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Feng Z., Hu W., de Stanchina E., Teresky A.K., Jin S., Lowe S., Levine A.J. The regulation of AMPK beta1, TSC2, and PTEN expression by p53: stress, cell and tissue specificity, and the role of these gene products in modulating the IGF-1-AKT-mTOR pathways. Cancer Res. 2007;67(7):3043–3053. doi: 10.1158/0008-5472.CAN-06-4149. [DOI] [PubMed] [Google Scholar]
- 124.Goiran T., Duplan E., Rouland L., El Manaa W., Lauritzen I., Dunys J., You H., Checler F., Alves da Costa C. Nuclear p53-mediated repression of autophagy involves PINK1 transcriptional down-regulation. Cell Death Differ. 2018;25(5):873–884. doi: 10.1038/s41418-017-0016-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Cenini G., Sultana R., Memo M., Butterfield D.A. Elevated levels of pro-apoptotic p53 and its oxidative modification by the lipid peroxidation product, HNE, in brain from subjects with amnestic mild cognitive impairment and Alzheimer’s disease. J. Cell. Mol. Med. 2008;12(3):987–994. doi: 10.1111/j.1582-4934.2008.00163.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Kitamura Y., Shimohama S., Kamoshima W., Matsuoka Y., Nomura Y., Taniguchi T. Changes of p53 in the brains of patients with Alzheimer’s disease. Biochem. Biophys. Res. Commun. 1997;232(2):418–421. doi: 10.1006/bbrc.1997.6301. [DOI] [PubMed] [Google Scholar]
- 127.Checler F., Dunys J., Pardossi-Piquard R., Alves da Costa C. p53 is regulated by and regulates members of the gamma-secretase complex. Neurodegener. Dis. 2010;7(1-3):50–55. doi: 10.1159/000283483. [DOI] [PubMed] [Google Scholar]
- 128.Hooper C., Meimaridou E., Tavassoli M., Melino G., Lovestone S., Killick R. p53 is upregulated in Alzheimer’s disease and induces tau phosphorylation in HEK293a cells. Neurosci. Lett. 2007;418(1):34–37. doi: 10.1016/j.neulet.2007.03.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Pattingre S., Tassa A., Qu X., Garuti R., Liang X.H., Mizushima N., Packer M., Schneider M.D., Levine B. Bcl-2 antiapoptotic proteins inhibit Beclin 1-dependent autophagy. Cell. 2005;122(6):927–939. doi: 10.1016/j.cell.2005.07.002. [DOI] [PubMed] [Google Scholar]
- 130.Michiorri S., Gelmetti V., Giarda E., Lombardi F., Romano F., Marongiu R., Nerini-Molteni S., Sale P., Vago R., Arena G., Torosantucci L., Cassina L., Russo M.A., Dallapiccola B., Valente E.M., Casari G. The Parkinson-associated protein PINK1 interacts with Beclin1 and promotes autophagy. Cell Death Differ. 2010;17(6):962–974. doi: 10.1038/cdd.2009.200. [DOI] [PubMed] [Google Scholar]
- 131.Lonskaya I., Hebron M.L., Desforges N.M., Franjie A., Moussa C.E. Tyrosine kinase inhibition increases functional parkin-Beclin-1 interaction and enhances amyloid clearance and cognitive performance. EMBO Mol. Med. 2013;5(8):1247–1262. doi: 10.1002/emmm.201302771. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Mandard S., Müller M., Kersten S. Peroxisome proliferator-activated receptor alpha target genes. Cell. Mol. Life Sci. 2004;61(4):393–416. doi: 10.1007/s00018-003-3216-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Lee J.M., Wagner M., Xiao R., Kim K.H., Feng D., Lazar M.A., Moore D.D. Nutrient-sensing nuclear receptors coordinate autophagy. Nature. 2014;516(7529):112–115. doi: 10.1038/nature13961. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Ghosh A., Jana M., Modi K., Gonzalez F.J., Sims K.B., Berry-Kravis E., Pahan K. Activation of peroxisome proliferator-activated receptor α induces lysosomal biogenesis in brain cells: implications for lysosomal storage disorders. J. Biol. Chem. 2015;290(16):10309–10324. doi: 10.1074/jbc.M114.610659. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Luo R., Su L.Y., Li G., Yang J., Liu Q., Yang L.X., Zhang D.F., Zhou H., Xu M., Fan Y., Li J., Yao Y.G. Activation of PPARA-mediated autophagy reduces Alzheimer disease-like pathology and cognitive decline in a murine model. Autophagy. 2020;6(1):52–69. doi: 10.1080/15548627.2019.1596488. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Chandra S., Roy A., Jana M., Pahan K. Cinnamic acid activates PPARα to stimulate Lysosomal biogenesis and lower Amyloid plaque pathology in an Alzheimer’s disease mouse model. Neurobiol. Dis. 2019;124:379–395. doi: 10.1016/j.nbd.2018.12.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Pajares M., Jiménez-Moreno N., García-Yagüe A.J., Escoll M., de Ceballos M.L., Van Leuven F., Rábano A., Yamamoto M., Rojo A.I., Cuadrado A. Transcription factor NFE2L2/NRF2 is a regulator of macroautophagy genes. Autophagy. 2016;12(10):1902–1916. doi: 10.1080/15548627.2016.1208889. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Hong S.J., Dawson T.M., Dawson V.L. Nuclear and mitochondrial conversations in cell death: PARP-1 and AIF signaling. Trends Pharmacol. Sci. 2004;25(5):259–264. doi: 10.1016/j.tips.2004.03.005. [DOI] [PubMed] [Google Scholar]
- 139.Kanninen K., Heikkinen R., Malm T., Rolova T., Kuhmonen S., Leinonen H., Ylä-Herttuala S., Tanila H., Levonen A.L., Koistinaho M., Koistinaho J. Intrahippocampal injection of a lentiviral vector expressing Nrf2 improves spatial learning in a mouse model of Alzheimer’s disease. Proc. Natl. Acad. Sci. USA. 2009;106(38):16505–16510. doi: 10.1073/pnas.0908397106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Rojo A.I., Pajares M., Rada P., Nuñez A., Nevado-Holgado A.J., Killik R., Van Leuven F., Ribe E., Lovestone S., Yamamoto M., Cuadrado A. NRF2 deficiency replicates transcriptomic changes in Alzheimer’s patients and worsens APP and TAU pathology. Redox Biol. 2017;13:444–451. doi: 10.1016/j.redox.2017.07.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Kim S., Lee D., Song J.C., Cho S.J., Yun S.M., Koh Y.H., Song J., Johnson G.V., Jo C. NDP52 associates with phosphorylated tau in brains of an Alzheimer disease mouse model. Biochem. Biophys. Res. Commun. 2014;454(1):196–201. doi: 10.1016/j.bbrc.2014.10.066. [DOI] [PubMed] [Google Scholar]
- 142.Jo C., Gundemir S., Pritchard S., Jin Y.N., Rahman I., Johnson G.V. Nrf2 reduces levels of phosphorylated tau protein by inducing autophagy adaptor protein NDP52. Nat. Commun. 2014;5:3496. doi: 10.1038/ncomms4496. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Cheng X., Shen D., Samie M., Xu H. Mucolipins: Intracellular TRPML1-3 channels. FEBS Lett. 2010;584(10):2013–2021. doi: 10.1016/j.febslet.2009.12.056. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Bae M., Patel N., Xu H., Lee M., Tominaga-Yamanaka K., Nath A., Geiger J., Gorospe M., Mattson M.P., Haughey N.J. Activation of TRPML1 clears intraneuronal Aβ in preclinical models of HIV infection. J. Neurosci. 2014;34(34):11485–11503. doi: 10.1523/JNEUROSCI.0210-14.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Grishchuk Y., Sri S., Rudinskiy N., Ma W., Stember K.G., Cottle M.W., Sapp E., Difiglia M., Muzikansky A., Betensky R.A., Wong A.M., Bacskai B.J., Hyman B.T., Kelleher R.J., III, Cooper J.D., Slaugenhaupt S.A. Behavioral deficits, early gliosis, dysmyelination and synaptic dysfunction in a mouse model of mucolipidosis IV. Acta Neuropathol. Commun. 2014;2:133. doi: 10.1186/s40478-014-0133-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Zhang L., Fang Y., Cheng X., Lian Y., Xu H., Zeng Z., Zhu H. TRPML1 Participates in the progression of Alzheimer’s Disease by regulating the PPARγ/AMPK/Mtor signalling pathway. Cell. Physiol. Biochem. 2017;43(6):2446–2456. doi: 10.1159/000484449. [DOI] [PubMed] [Google Scholar]
- 147.Fang E.F., Hou Y., Palikaras K., Adriaanse B.A., Kerr J.S., Yang B., Lautrup S., Hasan-Olive M.M., Caponio D., Dan X., Rocktäschel P., Croteau D.L., Akbari M., Greig N.H., Fladby T., Nilsen H., Cader M.Z., Mattson M.P., Tavernarakis N., Bohr V.A. Mitophagy inhibits amyloid-β and tau pathology and reverses cognitive deficits in models of Alzheimer’s disease. Nat. Neurosci. 2019;22(3):401–412. doi: 10.1038/s41593-018-0332-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Clark I.E., Dodson M.W., Jiang C., Cao J.H., Huh J.R., Seol J.H., Yoo S.J., Hay B.A., Guo M. Drosophila pink1 is required for mitochondrial function and interacts genetically with parkin. Nature. 2006;441(7097):1162–1166. doi: 10.1038/nature04779. [DOI] [PubMed] [Google Scholar]
- 149.Narendra D., Tanaka A., Suen D.F., Youle R.J. Parkin is recruited selectively to impaired mitochondria and promotes their autophagy. J. Cell Biol. 2008;183(5):795–803. doi: 10.1083/jcb.200809125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Jin S.M., Lazarou M., Wang C., Kane L.A., Narendra D.P., Youle R.J. Mitochondrial membrane potential regulates PINK1 import and proteolytic destabilization by PARL. J. Cell Biol. 2010;191(5):933–942. doi: 10.1083/jcb.201008084. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Takatori S., Ito G., Iwatsubo T. Cytoplasmic localization and proteasomal degradation of N-terminally cleaved form of PINK1. Neurosci. Lett. 2008;430(1):13–17. doi: 10.1016/j.neulet.2007.10.019. [DOI] [PubMed] [Google Scholar]
- 152.Vives-Bauza C., Zhou C., Huang Y., Cui M., de Vries R.L., Kim J., May J., Tocilescu M.A., Liu W., Ko H.S., Magrané J., Moore D.J., Dawson V.L., Grailhe R., Dawson T.M., Li C., Tieu K., Przedborski S. PINK1-dependent recruitment of Parkin to mitochondria in mitophagy. Proc. Natl. Acad. Sci. USA. 2010;107(1):378–383. doi: 10.1073/pnas.0911187107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Kazlauskaite A., Kondapalli C., Gourlay R., Campbell D.G., Ritorto M.S., Hofmann K., Alessi D.R., Knebel A., Trost M., Muqit M.M. Parkin is activated by PINK1-dependent phosphorylation of ubiquitin at Ser65. Biochem. J. 2014;460(1):127–139. doi: 10.1042/BJ20140334. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Kim Y., Park J., Kim S., Song S., Kwon S.K., Lee S.H., Kitada T., Kim J.M., Chung J. PINK1 controls mitochondrial localization of Parkin through direct phosphorylation. Biochem. Biophys. Res. Commun. 2008;377(3):975–980. doi: 10.1016/j.bbrc.2008.10.104. [DOI] [PubMed] [Google Scholar]
- 155.Banerjee K., Munshi S., Frank D.E., Gibson G.E. Abnormal glucose metabolism in alzheimer’s disease: relation to autophagy/mitophagy and therapeutic approaches. Neurochem. Res. 2015;40(12):2557–2569. doi: 10.1007/s11064-015-1631-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Du F., Yu Q., Yan S., Hu G., Lue L.F., Walker D.G., Wu L., Yan S.F., Tieu K., Yan S.S. PINK1 signalling rescues amyloid pathology and mitochondrial dysfunction in Alzheimer’s disease. Brain. 2017;140(12):3233–3251. doi: 10.1093/brain/awx258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Ye X., Sun X., Starovoytov V., Cai Q. Parkin-mediated mitophagy in mutant hAPP neurons and Alzheimer’s disease patient brains. Hum. Mol. Genet. 2015;24(10):2938–2951. doi: 10.1093/hmg/ddv056. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Hong X., Liu J., Zhu G., Zhuang Y., Suo H., Wang P., Huang D., Xu J., Huang Y., Yu M., Bian M., Sheng Z., Fei J., Song H., Behnisch T., Huang F. Parkin overexpression ameliorates hippocampal long-term potentiation and β-amyloid load in an Alzheimer’s disease mouse model. Hum. Mol. Genet. 2014;23(4):1056–1072. doi: 10.1093/hmg/ddt501. [DOI] [PubMed] [Google Scholar]
- 159.Harrison D.E., Strong R., Sharp Z.D., Nelson J.F., Astle C.M., Flurkey K., Nadon N.L., Wilkinson J.E., Frenkel K., Carter C.S., Pahor M., Javors M.A., Fernandez E., Miller R.A. Rapamycin fed late in life extends lifespan in genetically heterogeneous mice. Nature. 2009;460(7253):392–395. doi: 10.1038/nature08221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Boland B., Kumar A., Lee S., Platt F.M., Wegiel J., Yu W.H., Nixon R.A. Autophagy induction and autophagosome clearance in neurons: relationship to autophagic pathology in Alzheimer’s disease. J. Neurosci. 2008;28(27):6926–6937. doi: 10.1523/JNEUROSCI.0800-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.McGowan E., Pickford F., Kim J., Onstead L., Eriksen J., Yu C., Skipper L., Murphy M.P., Beard J., Das P., Jansen K., DeLucia M., Lin W.L., Dolios G., Wang R., Eckman C.B., Dickson D.W., Hutton M., Hardy J., Golde T. Abeta42 is essential for parenchymal and vascular amyloid deposition in mice. Neuron. 2005;47(2):191–199. doi: 10.1016/j.neuron.2005.06.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Polanco J.C., Li C., Durisic N., Sullivan R., Götz J. Exosomes taken up by neurons hijack the endosomal pathway to spread to interconnected neurons. Acta Neuropathol. Commun. 2018;6(1):10. doi: 10.1186/s40478-018-0514-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Soll C., Clavien P.A. Inhibition of mammalian target of rapamycin: the janus face of immunosuppression? Hepatology. 2010;51(4):1113–1115. doi: 10.1002/hep.23582. [DOI] [PubMed] [Google Scholar]
- 164.Thellung S., Corsaro A., Nizzari M., Barbieri F., Florio T. Autophagy Activator Drugs: A new opportunity in neuroprotection from misfolded protein toxicity. Int. J. Mol. Sci. 2019;20(4):E901. doi: 10.3390/ijms20040901. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Vlad S.C., Miller D.R., Kowall N.W., Felson D.T. Protective effects of NSAIDs on the development of Alzheimer disease. Neurology. 2008;70(19):1672–1677. doi: 10.1212/01.wnl.0000311269.57716.63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Carreras I., McKee A.C., Choi J.K., Aytan N., Kowall N.W., Jenkins B.G., Dedeoglu A. R-flurbiprofen improves tau, but not Aß pathology in a triple transgenic model of Alzheimer’s disease. Brain Res. 2013;1541:115–127. doi: 10.1016/j.brainres.2013.10.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Chandra S., Jana M., Pahan K. Aspirin induces lysosomal biogenesis and attenuates amyloid plaque pathology in a mouse model of Alzheimer’s Disease via PPARα. J. Neurosci. 2018;38(30):6682–6699. doi: 10.1523/JNEUROSCI.0054-18.2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Ayyadevara S., Balasubramaniam M., Kakraba S., Alla R., Mehta J.L., Shmookler Reis R.J. Aspirin-mediated acetylation protects against multiple neurodegenerative pathologies by impeding protein aggregation. Antioxid. Redox Signal. 2017;27(17):1383–1396. doi: 10.1089/ars.2016.6978. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Matsunaga S., Kishi T., Annas P., Basun H., Hampel H., Iwata N. Lithium as a treatment for Alzheimer’s Disease: A systematic review and meta-analysis. J. Alzheimers Dis. 2015;48(2):403–410. doi: 10.3233/JAD-150437. [DOI] [PubMed] [Google Scholar]
- 170.Forlenza O.V., Diniz B.S., Radanovic M., Santos F.S., Talib L.L., Gattaz W.F. Disease-modifying properties of long-term lithium treatment for amnestic mild cognitive impairment: randomised controlled trial. Br. J. Psychiatry. 2011;198(5):351–356. doi: 10.1192/bjp.bp.110.080044. [DOI] [PubMed] [Google Scholar]
- 171.Yang J., Takahashi Y., Cheng E., Liu J., Terranova P.F., Zhao B., Thrasher J.B., Wang H.G., Li B. GSK-3beta promotes cell survival by modulating Bif-1-dependent autophagy and cell death. J. Cell Sci. 2010;123(Pt 6):861–870. doi: 10.1242/jcs.060475. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Sarkar S., Floto R.A., Berger Z., Imarisio S., Cordenier A., Pasco M., Cook L.J., Rubinsztein D.C. Lithium induces autophagy by inhibiting inositol monophosphatase. J. Cell Biol. 2005;170(7):1101–1111. doi: 10.1083/jcb.200504035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Grossberg G.T., Pejović V., Miller M.L., Graham S.M. Memantine therapy of behavioral symptoms in community-dwelling patients with moderate to severe Alzheimer’s disease. Dement. Geriatr. Cogn. Disord. 2009;27(2):164–172. doi: 10.1159/000200013. [DOI] [PubMed] [Google Scholar]
- 174.Hirano K., Fujimaki M., Sasazawa Y., Yamaguchi A., Ishikawa K.I., Miyamoto K., Souma S., Furuya N., Imamichi Y., Yamada D., Saya H., Akamatsu W., Saiki S., Hattori N. Neuroprotective effects of memantine via enhancement of autophagy. Biochem. Biophys. Res. Commun. 2019;518(1):161–170. doi: 10.1016/j.bbrc.2019.08.025. [DOI] [PubMed] [Google Scholar]
- 175.Sestito S., Daniele S., Pietrobono D., Citi V., Bellusci L., Chiellini G., Calderone V., Martini C., Rapposelli S. Memantine prodrug as a new agent for Alzheimer’s Disease. Sci. Rep. 2019;9(1):4612. doi: 10.1038/s41598-019-40925-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Reiter R.J. The pineal gland and melatonin in relation to aging: a summary of the theories and of the data. Exp. Gerontol. 1995;30(3-4):199–212. doi: 10.1016/0531-5565(94)00045-5. [DOI] [PubMed] [Google Scholar]
- 177.Liu R.Y., Zhou J.N., van Heerikhuize J., Hofman M.A., Swaab D.F. Decreased melatonin levels in postmortem cerebrospinal fluid in relation to aging, Alzheimer’s disease, and apolipoprotein E-epsilon4/4 genotype. J. Clin. Endocrinol. Metab. 1999;84(1):323–327. doi: 10.1210/jcem.84.1.5394. [DOI] [PubMed] [Google Scholar]
- 178.Chang H.M., Wu U.I., Lan C.T. Melatonin preserves longevity protein (sirtuin 1) expression in the hippocampus of total sleep-deprived rats. J. Pineal Res. 2009;47(3):211–220. doi: 10.1111/j.1600-079X.2009.00704.x. [DOI] [PubMed] [Google Scholar]
- 179.Cristòfol R., Porquet D., Corpas R., Coto-Montes A., Serret J., Camins A., Pallàs M., Sanfeliu C. Neurons from senescence-accelerated SAMP8 mice are protected against frailty by the sirtuin 1 promoting agents melatonin and resveratrol. J. Pineal Res. 2012;52(3):271–281. doi: 10.1111/j.1600-079X.2011.00939.x. [DOI] [PubMed] [Google Scholar]
- 180.Luengo E., Buendia I., Fernández-Mendívil C., Trigo-Alonso P., Negredo P., Michalska P., Hernández-García B., Sánchez-Ramos C., Bernal J.A., Ikezu T., León R., López M.G. Pharmacological doses of melatonin impede cognitive decline in tau-related Alzheimer models, once tauopathy is initiated, by restoring the autophagic flux. J. Pineal Res. 2019;67(1):e12578. doi: 10.1111/jpi.12578. [DOI] [PubMed] [Google Scholar]
- 181.Caton P.W., Nayuni N.K., Kieswich J., Khan N.Q., Yaqoob M.M., Corder R. Metformin suppresses hepatic gluconeogenesis through induction of SIRT1 and GCN5. J. Endocrinol. 2010;205(1):97–106. doi: 10.1677/JOE-09-0345. [DOI] [PubMed] [Google Scholar]
- 182.Song Y.M., Song S.O., Jung Y.K., Kang E.S., Cha B.S., Lee H.C., Lee B.W. Dimethyl sulfoxide reduces hepatocellular lipid accumulation through autophagy induction. Autophagy. 2012;8(7):1085–1097. doi: 10.4161/auto.20260. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Song Y.M., Lee W.K., Lee Y.H., Kang E.S., Cha B.S., Lee B.W. Metformin Restores Parkin-Mediated Mitophagy, Suppressed by Cytosolic p53. Int. J. Mol. Sci. 2016;17(1):E122. doi: 10.3390/ijms17010122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Ng T.P., Feng L., Yap K.B., Lee T.S., Tan C.H., Winblad B. Long-term metformin usage and cognitive function among older adults with diabetes. J. Alzheimers Dis. 2014;41(1):61–68. doi: 10.3233/JAD-131901. [DOI] [PubMed] [Google Scholar]
- 185.Hsu C.C., Wahlqvist M.L., Lee M.S., Tsai H.N. Incidence of dementia is increased in type 2 diabetes and reduced by the use of sulfonylureas and metformin. J. Alzheimers Dis. 2011;24(3):485–493. doi: 10.3233/JAD-2011-101524. [DOI] [PubMed] [Google Scholar]
- 186.Chen J.L., Luo C., Pu D., Zhang G.Q., Zhao Y.X., Sun Y., Zhao K.X., Liao Z.Y., Lv A.K., Zhu S.Y., Zhou J., Xiao Q. Metformin attenuates diabetes-induced tau hyperphosphorylation in vitro and in vivo by enhancing autophagic clearance. Exp. Neurol. 2019;311:44–56. doi: 10.1016/j.expneurol.2018.09.008. [DOI] [PubMed] [Google Scholar]
- 187.Son S.M., Shin H.J., Byun J., Kook S.Y., Moon M., Chang Y.J., Mook-Jung I. Metformin facilitates amyloid-β generation by β- and γ-secretases via autophagy activation. J. Alzheimers Dis. 2016;51(4):1197–1208. doi: 10.3233/JAD-151200. [DOI] [PubMed] [Google Scholar]
- 188.Imfeld P., Bodmer M., Jick S.S., Meier C.R. Metformin, other antidiabetic drugs, and risk of Alzheimer’s disease: a population-based case-control study. J. Am. Geriatr. Soc. 2012;60(5):916–921. doi: 10.1111/j.1532-5415.2012.03916.x. [DOI] [PubMed] [Google Scholar]
- 189.Littlejohns T.J., Henley W.E., Lang I.A., Annweiler C., Beauchet O., Chaves P.H., Fried L., Kestenbaum B.R., Kuller L.H., Langa K.M., Lopez O.L., Kos K., Soni M., Llewellyn D.J. Vitamin D and the risk of dementia and Alzheimer disease. Neurology. 2014;83(10):920–928. doi: 10.1212/WNL.0000000000000755. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Tavera-Mendoza L.E., Westerling T., Libby E., Marusyk A., Cato L., Cassani R., Cameron L.A., Ficarro S.B., Marto J.A., Klawitter J., Brown M. Vitamin D receptor regulates autophagy in the normal mammary gland and in luminal breast cancer cells. Proc. Natl. Acad. Sci. USA. 2017;114(11):E2186–E2194. doi: 10.1073/pnas.1615015114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Guo Y.X., He L.Y., Zhang M., Wang F., Liu F., Peng W.X. 1,25-Dihydroxyvitamin D3 regulates expression of LRP1 and RAGE in vitro and in vivo, enhancing Aβ1-40 brain-to-blood efflux and peripheral uptake transport. Neuroscience. 2016;322:28–38. doi: 10.1016/j.neuroscience.2016.01.041. [DOI] [PubMed] [Google Scholar]
- 192.Durk M.R., Han K., Chow E.C., Ahrens R., Henderson J.T., Fraser P.E., Pang K.S. 1α,25-Dihydroxyvitamin D3 reduces cerebral amyloid-β accumulation and improves cognition in mouse models of Alzheimer’s disease. J. Neurosci. 2014;34(21):7091–7101. doi: 10.1523/JNEUROSCI.2711-13.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Høyer-Hansen M., Bastholm L., Mathiasen I.S., Elling F., Jäättelä M. Vitamin D analog EB1089 triggers dramatic lysosomal changes and Beclin 1-mediated autophagic cell death. Cell Death Differ. 2005;12(10):1297–1309. doi: 10.1038/sj.cdd.4401651. [DOI] [PubMed] [Google Scholar]
- 194.Høyer-Hansen M., Bastholm L., Szyniarowski P., Campanella M., Szabadkai G., Farkas T., Bianchi K., Fehrenbacher N., Elling F., Rizzuto R., Mathiasen I.S., Jäättelä M. Control of macroautophagy by calcium, calmodulin-dependent kinase kinase-beta, and Bcl-2. Mol. Cell. 2007;25(2):193–205. doi: 10.1016/j.molcel.2006.12.009. [DOI] [PubMed] [Google Scholar]
- 195.Chen X., Li M., Li L., Xu S., Huang D., Ju M., Huang J., Chen K., Gu H. Trehalose, sucrose and raffinose are novel activators of autophagy in human keratinocytes through an mTOR-independent pathway. Sci. Rep. 2016;6:28423. doi: 10.1038/srep28423. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Du J., Liang Y., Xu F., Sun B., Wang Z. Trehalose rescues Alzheimer’s disease phenotypes in APP/PS1 transgenic mice. J. Pharm. Pharmacol. 2013;65(12):1753–1756. doi: 10.1111/jphp.12108. [DOI] [PubMed] [Google Scholar]
- 197.Schaeffer V., Lavenir I., Ozcelik S., Tolnay M., Winkler D.T., Goedert M. Stimulation of autophagy reduces neurodegeneration in a mouse model of human tauopathy. Brain. 2012;135(Pt 7):2169–2177. doi: 10.1093/brain/aws143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198.Vingtdeux V., Giliberto L., Zhao H., Chandakkar P., Wu Q., Simon J.E., Janle E.M., Lobo J., Ferruzzi M.G., Davies P., Marambaud P. AMP-activated protein kinase signaling activation by resveratrol modulates amyloid-beta peptide metabolism. J. Biol. Chem. 2010;285(12):9100–9113. doi: 10.1074/jbc.M109.060061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199.Armour S.M., Baur J.A., Hsieh S.N., Land-Bracha A., Thomas S.M., Sinclair D.A. Inhibition of mammalian S6 kinase by resveratrol suppresses autophagy. Aging (Albany NY) 2009;1(6):515–528. doi: 10.18632/aging.100056. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.Yu K.C., Kwan P., Cheung S.K.K., Ho A., Baum L. Effects of resveratrol and morin on insoluble tau in tau transgenic mice. Transl. Neurosci. 2018;9:54–60. doi: 10.1515/tnsci-2018-0010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Karimipour M., Rahbarghazi R., Tayefi H., Shimia M., Ghanadian M., Mahmoudi J., Bagheri H.S. Quercetin promotes learning and memory performance concomitantly with neural stem/progenitor cell proliferation and neurogenesis in the adult rat dentate gyrus. Int. J. Dev. Neurosci. 2019;74:18–26. doi: 10.1016/j.ijdevneu.2019.02.005. [DOI] [PubMed] [Google Scholar]
- 202.Jiménez-Aliaga K., Bermejo-Bescós P., Benedí J., Martín-Aragón S. Quercetin and rutin exhibit antiamyloidogenic and fibril-disaggregating effects in vitro and potent antioxidant activity in APPswe cells. Life Sci. 2011;89(25-26):939–945. doi: 10.1016/j.lfs.2011.09.023. [DOI] [PubMed] [Google Scholar]
- 203.Regitz C., Dussling L.M., Wenzel U. Amyloid-beta (Abeta(1)(-)(4)(2))-induced paralysis in Caenorhabditis elegans is inhibited by the polyphenol quercetin through activation of protein degradation pathways. Mol. Nutr. Food Res. 2014;58(10):1931–1940. doi: 10.1002/mnfr.201400014. [DOI] [PubMed] [Google Scholar]
- 204.Li Y., Tian Q., Li Z., Dang M., Lin Y., Hou X. Activation of Nrf2 signaling by sitagliptin and quercetin combination against β-amyloid induced Alzheimer’s disease in rats. Drug Dev. Res. 2019;80(6):837–845. doi: 10.1002/ddr.21567. [DOI] [PubMed] [Google Scholar]
- 205.Suganthy N., Devi K.P., Nabavi S.F., Braidy N., Nabavi S.M. Bioactive effects of quercetin in the central nervous system: Focusing on the mechanisms of actions. Biomed. Pharmacother. 2016;84:892–908. doi: 10.1016/j.biopha.2016.10.011. [DOI] [PubMed] [Google Scholar]
- 206.Liu Y., Zhou H., Yin T., Gong Y., Yuan G., Chen L., Liu J. Quercetin-modified gold-palladium nanoparticles as a potential autophagy inducer for the treatment of Alzheimer’s disease. J. Colloid Interface Sci. 2019;552:388–400. doi: 10.1016/j.jcis.2019.05.066. [DOI] [PubMed] [Google Scholar]
- 207.Guo J., Chang L., Zhang X., Pei S., Yu M., Gao J. Ginsenoside compound K promotes β-amyloid peptide clearance in primary astrocytes via autophagy enhancement. Exp. Ther. Med. 2014;8(4):1271–1274. doi: 10.3892/etm.2014.1885. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208.Yang Q., Lin J., Zhang H., Liu Y., Kan M., Xiu Z., Chen X., Lan X., Li X., Shi X., Li N., Qu X. Ginsenoside compound k regulates amyloid β via the Nrf2/Keap1 signaling pathway in mice with scopolamine hydrobromide-induced memory impairments. J. Mol. Neurosci. 2019;67(1):62–71. doi: 10.1007/s12031-018-1210-3. [DOI] [PubMed] [Google Scholar]
- 209.Yao X.C., Xue X., Zhang H.T., Zhu M.M., Yang X.W., Wu C.F., Yang J.Y. Pseudoginsenoside-F11 alleviates oligomeric β-amyloid-induced endosome-lysosome defects in microglia. Traffic. 2019;20(1):61–70. doi: 10.1111/tra.12620. [DOI] [PubMed] [Google Scholar]
- 210.Song X.Y., Hu J.F., Chu S.F., Zhang Z., Xu S., Yuan Y.H., Han N., Liu Y., Niu F., He X., Chen N.H. Ginsenoside Rg1 attenuates okadaic acid induced spatial memory impairment by the GSK3β/tau signaling pathway and the Aβ formation prevention in rats. Eur. J. Pharmacol. 2013;710(1-3):29–38. doi: 10.1016/j.ejphar.2013.03.051. [DOI] [PubMed] [Google Scholar]
- 211.Hishikawa N., Takahashi Y., Amakusa Y., Tanno Y., Tuji Y., Niwa H., Murakami N., Krishna U.K. Effects of turmeric on Alzheimer’s disease with behavioral and psychological symptoms of dementia. Ayu. 2012;33(4):499–504. doi: 10.4103/0974-8520.110524. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212.Zhang L., Fang Y., Cheng X., Lian Y., Zeng Z., Wu C., Zhu H., Xu H. The potential protective effect of curcumin on amyloid-β-42 induced cytotoxicity in HT-22 Cells. BioMed Res. Int. 2018;2018:8134902. doi: 10.1155/2018/8134902. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213.Wang C., Zhang X., Teng Z., Zhang T., Li Y. Downregulation of PI3K/Akt/mTOR signaling pathway in curcumin-induced autophagy in APP/PS1 double transgenic mice. Eur. J. Pharmacol. 2014;740:312–320. doi: 10.1016/j.ejphar.2014.06.051. [DOI] [PubMed] [Google Scholar]
- 214.Anand P., Kunnumakkara A.B., Newman R.A., Aggarwal B.B. Bioavailability of curcumin: problems and promises. Mol. Pharm. 2007;4(6):807–818. doi: 10.1021/mp700113r. [DOI] [PubMed] [Google Scholar]
- 215.Song J.X., Sun Y.R., Peluso I., Zeng Y., Yu X., Lu J.H., Xu Z., Wang M.Z., Liu L.F., Huang Y.Y., Chen L.L., Durairajan S.S., Zhang H.J., Zhou B., Zhang H.Q., Lu A., Ballabio A., Medina D.L., Guo Z., Li M. A novel curcumin analog binds to and activates TFEB in vitro and in vivo independent of MTOR inhibition. Autophagy. 2016;12(8):1372–1389. doi: 10.1080/15548627.2016.1179404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216.Bagherniya M., Butler A.E., Barreto G.E., Sahebkar A. The effect of fasting or calorie restriction on autophagy induction: A review of the literature. Ageing Res. Rev. 2018;47:183–197. doi: 10.1016/j.arr.2018.08.004. [DOI] [PubMed] [Google Scholar]
- 217.Rickenbacher A., Jang J.H., Limani P., Ungethüm U., Lehmann K., Oberkofler C.E., Weber A., Graf R., Humar B., Clavien P.A. Fasting protects liver from ischemic injury through Sirt1-mediated downregulation of circulating HMGB1 in mice. J. Hepatol. 2014;61(2):301–308. doi: 10.1016/j.jhep.2014.04.010. [DOI] [PubMed] [Google Scholar]
- 218.Golbidi S., Daiber A., Korac B., Li H., Essop M.F., Laher I. Health benefits of fasting and caloric restriction. Curr. Diab. Rep. 2017;17(12):123. doi: 10.1007/s11892-017-0951-7. [DOI] [PubMed] [Google Scholar]
- 219.Halagappa V.K., Guo Z., Pearson M., Matsuoka Y., Cutler R.G., Laferla F.M., Mattson M.P. Intermittent fasting and caloric restriction ameliorate age-related behavioral deficits in the triple-transgenic mouse model of Alzheimer’s disease. Neurobiol. Dis. 2007;26(1):212–220. doi: 10.1016/j.nbd.2006.12.019. [DOI] [PubMed] [Google Scholar]
- 220.Ntsapi C., Loos B. Caloric restriction and the precision-control of autophagy: A strategy for delaying neurodegenerative disease progression. Exp. Gerontol. 2016;83:97–111. doi: 10.1016/j.exger.2016.07.014. [DOI] [PubMed] [Google Scholar]
- 221.Chen X., Kondo K., Motoki K., Homma H., Okazawa H. Fasting activates macroautophagy in neurons of Alzheimer’s disease mouse model but is insufficient to degrade amyloid-beta. Sci. Rep. 2015;5:12115. doi: 10.1038/srep12115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222.Gregosa A., Vinuesa Á., Todero M.F., Pomilio C., Rossi S.P., Bentivegna M., Presa J., Wenker S., Saravia F., Beauquis J. Periodic dietary restriction ameliorates amyloid pathology and cognitive impairment in PDAPP-J20 mice: Potential implication of glial autophagy. Neurobiol. Dis. 2019;132:104542. doi: 10.1016/j.nbd.2019.104542. [DOI] [PubMed] [Google Scholar]
- 223.Hadem I.K.H., Sharma R. Differential regulation of hippocampal IGF-1-Associated signaling proteins by dietary restriction in aging mouse. Cell. Mol. Neurobiol. 2017;37(6):985–993. doi: 10.1007/s10571-016-0431-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 224.Qin W., Yang T., Ho L., Zhao Z., Wang J., Chen L., Zhao W., Thiyagarajan M., MacGrogan D., Rodgers J.T., Puigserver P., Sadoshima J., Deng H., Pedrini S., Gandy S., Sauve A.A., Pasinetti G.M. Neuronal SIRT1 activation as a novel mechanism underlying the prevention of Alzheimer disease amyloid neuropathology by calorie restriction. J. Biol. Chem. 2006;281(31):21745–21754. doi: 10.1074/jbc.M602909200. [DOI] [PubMed] [Google Scholar]
- 225.Groot C., Hooghiemstra A.M., Raijmakers P.G., van Berckel B.N., Scheltens P., Scherder E.J., van der Flier W.M., Ossenkoppele R. The effect of physical activity on cognitive function in patients with dementia: A meta-analysis of randomized control trials. Ageing Res. Rev. 2016;25:13–23. doi: 10.1016/j.arr.2015.11.005. [DOI] [PubMed] [Google Scholar]
- 226.Hoffmann K., Sobol N.A., Frederiksen K.S., Beyer N., Vogel A., Vestergaard K., Brændgaard H., Gottrup H., Lolk A., Wermuth L., Jacobsen S., Laugesen L.P., Gergelyffy R.G., Høgh P., Bjerregaard E., Andersen B.B., Siersma V., Johannsen P., Cotman C.W., Waldemar G., Hasselbalch S.G. Moderate-to-high intensity physical exercise in patients with alzheimer’s disease: a randomized controlled trial. J. Alzheimers Dis. 2016;50(2):443–453. doi: 10.3233/JAD-150817. [DOI] [PubMed] [Google Scholar]
- 227.Larson E.B., Wang L., Bowen J.D., McCormick W.C., Teri L., Crane P., Kukull W. Exercise is associated with reduced risk for incident dementia among persons 65 years of age and older. Ann. Intern. Med. 2006;144(2):73–81. doi: 10.7326/0003-4819-144-2-200601170-00004. [DOI] [PubMed] [Google Scholar]
- 228.Kou X., Chen D., Chen N. Physical activity alleviates cognitive dysfunction of alzheimer’s disease through regulating the mTOR signaling pathway. Int. J. Mol. Sci. 2019;20(7):E1591. doi: 10.3390/ijms20071591. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 229.Herring A., Münster Y., Metzdorf J., Bolczek B., Krüssel S., Krieter D., Yavuz I., Karim F., Roggendorf C., Stang A., Wang Y., Hermann D.M., Teuber-Hanselmann S., Keyvani K. Late running is not too late against Alzheimer’s pathology. Neurobiol. Dis. 2016;94:44–54. doi: 10.1016/j.nbd.2016.06.003. [DOI] [PubMed] [Google Scholar]
- 230.Liu W., Wang Z., Xia Y., Kuang H., Liu S., Li L., Tang C., Yin D. The balance of apoptosis and autophagy via regulation of the AMPK signal pathway in aging rat striatum during regular aerobic exercise. Exp. Gerontol. 2019;124:110647. doi: 10.1016/j.exger.2019.110647. [DOI] [PubMed] [Google Scholar]
- 231.Smith P.J., Blumenthal J.A., Hoffman B.M., Cooper H., Strauman T.A., Welsh-Bohmer K., Browndyke J.N., Sherwood A. Aerobic exercise and neurocognitive performance: a meta-analytic review of randomized controlled trials. Psychosom. Med. 2010;72(3):239–252. doi: 10.1097/PSY.0b013e3181d14633. [DOI] [PMC free article] [PubMed] [Google Scholar]



