Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2024 May 24.
Published in final edited form as: J Alzheimers Dis. 2024;99(2):447–470. doi: 10.3233/JAD-240042

Evidence that Alzheimer’s disease is a disease of competitive synaptic plasticity gone awry

Zhen Huang 1
PMCID: PMC11119021  NIHMSID: NIHMS1992046  PMID: 38669548

Abstract

Mounting evidence indicates that a physiological function of amyloid β (Aβ) is to mediate neural activity-dependent homeostatic and competitive synaptic plasticity in the brain. I have previously summarized the lines of evidence supporting this hypothesis and highlighted the similarities between Aβ and anti-microbial peptides in mediating cell/synapse competition. In cell competition, anti-microbial peptides deploy a multitude of mechanisms to ensure both self-protection and competitor elimination. Here I review recent studies showing that similar mechanisms are at play in Aβ-mediated synapse competition and perturbations in these mechanisms underpin Alzheimer’s disease. Specifically, I discuss evidence that Aβ and ApoE, two crucial players in Alzheimer’s disease, co-operate in the regulation of synapse competition. Glial ApoE promotes self-protection by increasing the production of trophic monomeric Aβ and inhibiting its assembly into toxic oligomers. Conversely, Aβ oligomers, once assembled, promote the elimination of competitor synapses via direct toxic activity and amplification of “eat-me” signals promoting the elimination of weak synapses. I further summarize evidence that neuronal ApoE may be part of a gene regulatory network that normally promotes competitive plasticity, explaining the selective vulnerability of ApoE expressing neurons in Alzheimer’s brains. Lastly, I discuss evidence that sleep may be key to Aβ-orchestrated plasticity, in which sleep is not only induced by Aβ but is also required for Aβ-mediated plasticity, underlining the link between sleep and Alzheimer’s disease. Together, these results strongly argue that Alzheimer’s disease is a disease of competitive synaptic plasticity gone awry, a novel perspective that may promote Alzheimer’s disease research.

Keywords: Alzheimer’s disease, amyloid beta, amyloid precursor protein, DNA damage repair, synaptic competition, homeostatic plasticity, competitive plasticity, ApoE, dendritic spine, cellular prion protein, mGluR5, phosphatidylserine, sleep

Introduction

Aβ is the main component of amyloid plaques found in the brains of Alzheimer’s disease, a neurodegenerative condition affecting tens of millions of people worldwide. The invariable association of Aβ pathology with Alzheimer’s disease and the discovery that genetic mutations affecting Aβ production predictively result in the development of early-onset Alzheimer’s disease (also known as familial Alzheimer’s disease) strongly indicate that Aβ likely plays a central role in Alzheimer’s disease pathogenesis. Decades of research into the activity and regulation of Aβ also suggest that it plays a physiological role in the healthy brain. Based on many converging lines of evidence from several different fields, I have proposed that a physiological function of Aβ in the brain may be to mediate neural activity-dependent homeostatic and competitive synaptic plasticity, including activity-dependent synapse competition [1]. In this process, Aβ acts not only as a reporter of circuit-level neural activity in the brain but also as an activity level-dependent effector that directly mediates homeostatic and competitive synaptic plasticity. I propose that Aβ remains as monomer-related neuroprotective species when circuit-level activity and Aβ concentration are both low, but forms toxic oligomeric species when high-level circuit activity increases Aβ production and concentration, which then lead to synaptic competition and the weakening or even elimination of weak synapses. In prototypical cell competition, besides the action of anti-microbial peptides the similarities of which to Aβ I have previously reviewed, many other mechanisms are also activated in both the steps of self-protection and the steps of competitor killing [1]. Recent studies show that similar mechanisms, including the regulation of Aβ monomer production and assembly into oligomers by astrocytic ApoE [224] and the amplification of “eat-me” signals displayed at weak synapses by Aβ oligomers [2541], may also be at work during neural activity-dependent axon/synapse competition. These studies also show that sleep is induced by Aβ oligomer injection into the brain and crucial to both homeostatic and competitive synaptic plasticity [4254], potentially explaining the link between sleep disturbance and Alzheimer’s’ disease. In this article, I will review these findings and discuss how they have shed new light on the physiological and pathological functions of Aβ, ApoE, and other genes in the healthy and the Alzheimer’s disease brain and how they have provided new insight into the regulation of normal brain synaptic plasticity as well as the pathogenesis of Alzheimer’s disease.

Relationship between roles played by ApoE and AICD in competing synapse self-protection and in Alzheimer’s disease

In cell competition, competing cells activating pathways to kill competitors must also at the same time protect themselves from the activity of these same pathways. In the simplest form of cell competition such as anti-microbial peptide-mediated competition, bacterial cells engaging competition are known to up-regulate the expression of the so-called immunity proteins, which, both membrane-bound and secreted, can bind to anti-microbial peptides and impede their aggregation into toxic oligomers in the immediate vicinity of these cells [1]. If Aβ acts similarly in axon/synapse competition, similar mechanisms may also exist for protection against toxic Aβ species. Indeed, a large body of evidence indicates that both ApoE and amyloid precursor protein (APP), likely among others, are involved in self-protection during axon/synapse competition [224, 5567]. For example, studies show that ApoE promotes Aβ degradation and suppresses Aβ oligomerization [224] and that APP intracellular cleavage products promote mechanisms that repairs DNA damage induced by reactive oxygen species and neural activity during axon/synapse competition [5567]. Furthermore, compromised functions in these processes have been linked to increased risk to Alzheimer’s disease. Below, in this section, I will discuss these results and highlight how these findings help unifying our understanding of the functions of Aβ, APP, and ApoE under physiological and pathological conditions.

Regulation of Aβ oligomerization and degradation by ApoE and its role in Alzheimer’s disease

ApoE is an extracellular lipid-binding protein with prominent roles in brain lipid metabolism [68, 69] and the ApoE4 allele is one of the highest genetic risk factors in late-onset Alzheimer’s disease development [70, 71]. Several lines of evidence indicate that ApoE proteins, including those induced in neurons during heightened neural activity, may participate in axon/synapse competition by regulating both neuronal self-protection and competitor elimination (for roles of ApoE in competitor elimination, see discussion in the next section) [7277]. First, ApoE shows several properties and activities that befit such a role. For example, ApoE expression is induced in cortical neurons by glutamate [72], consistent with its regulation by neural activity. In the brain, while ApoE is well known to be most highly expressed in astrocytes, under high activity conditions such as kainic acid treatment, strong ApoE expression is also induced in neurons [73, 74]. In vitro, ApoE-containing lipoproteins protect neurons from receptor pathway-mediated apoptosis [75]. In vivo, neuronal expression of ApoE3 (but not ApoE4) also protects neurons from excitotoxic injury [76]. Furthermore, human ApoE expression in C. elegans significantly attenuates Aβ-induced neurodegeneration [77]. All these findings support the interpretation that ApoE may play a protective role in Aβ-mediated axon/synapse competition. (A note on the ApoE4 allele: while known to be the highest genetic risk factor for late-onset Alzheimer’s disease [70, 71], studies show that this allele may have allele-specific effects on several processes related to axon/synapse competition, including, for example, APP transcription and Aβ secretion [21], synaptogenesis [24], and NMDA receptor activity [78]. Some studies suggest that ApoE4 may have an enhancing effect on brain functions such as visual working memory [79], while other studies show that it impairs neuronal insulin signaling [80] and leads to the generation of allele-specific toxic proteolytic products [81]. As such, it is at present unclear if the ApoE4 mutation is purely pathological or may potentially confer advantage in specific aspects of brain function).

Several potential mechanisms may explain the protective effects observed of ApoE (Fig. 1). First, at the level of direct physical interactions, ApoE has been found to strongly influence the process of Aβ aggregation and as a result the amount of toxic Aβ species that may be produced. For example, early studies show that purified ApoE binds to Aβ peptides with significant affinity [26] (but also [7]). Recent investigations into the effects of ApoE on the different phases of Aβ oligomerization further show that it likely has differential effects on the production rate of different species of Aβ associated with different levels of toxicity. For example, studies analyzing the kinetics of Aβ oligomerization have consistently shown that purified ApoE slows down the growth of toxic Aβ oligomer species [810]. An EM study similarly shows that ApoE prevents Aβ oligomer formation [11]. Other studies show that ApoE may promote the formation of amyloid filaments or high-molecular-weight oligomers that are less toxic [1215]. In addition, ApoE-related clusterin has also been found to similarly sequester Aβ oligomers [82]. Together, these results suggest that ApoE may potentially play a neuroprotective role during Aβ-mediated axon/synapse competition. Interestingly, of the different ApoE alleles, ApoE4 appears to have a high level of activity in stabilizing soluble toxic Aβ oligomers [8, 10], which may contribute to the increased risk of Alzheimer’s disease development associated with this allele.

Figure 1. Mechanisms by which astrocytic ApoE may provide protection during Aβ-mediated axon/synapse competition.

Figure 1.

A. Several studies (see text for details) show that ApoE may both inhibit the formation of toxic Aβ species, potentially at steps of both primary and secondary nucleation, and promote the formation of less toxic high-molecular -weight (HMW) Aβ oligomers. B. There is also evidence indicating that ApoE may inhibit the transcription and secretion of inflammatory cytokines as well as promote phagocytosis and Aβ degradation by both activating cell surface receptors and promoting cholesterol efflux. Figure created with BioRender.com.

In vivo, the effects of manipulating ApoE levels on Aβ are more complex and harder to interpret and may include both direct and indirect effects (see also below on roles of ApoE in glial phagocytosis and Aβ clearance). Nonetheless, several studies show that ApoE appears to decrease Aβ toxicity in vivo [1620]. For example, early studies show that genetic removal of all ApoE from the brain appears to drastically reduce the total amounts of amyloid plaques from an APP overexpression mouse brain [16], while recent studies show that these manipulations have stage-specific effects on amyloid pathology [17, 18]. Most notably, new studies show that a key effect of ApoE removal in vivo may be in the reduction of amyloid plaque compaction, an effect that may be deleterious since it may result in increased amounts of diffuse toxic oligomers [19]. Other studies also show that genetic removal of all ApoE from APP overexpression mice leads to elevated levels of soluble Aβ in the brain [20]. Altogether, in vitro and in vivo findings suggest that ApoE may potentially serve neuroprotective roles in activity-induced axon/synapse competition by physically interacting with Aβ and modulating the kinetics of Aβ oligomer and other aggregate formation. Besides these lines of evidence, ApoE has also been found to specifically promote in human neurons the transcription of APP and the secretion of Aβ monomers [21] (A significant number of studies show that Aβ monomers and low-concentration oligomers are neuroprotective and positively regulate synapse formation, function as well as learning and memory [8394]). ApoE also increases APP interaction with β- and γ-secretases and promotes Aβ production through regulating cholesterol [22, 23]. Thus, ApoE may potentially protect competing axons/synapses additionally through up-regulating Aβ monomer levels. Lastly, ApoE has been found to promote synaptogenesis through CREB activity independent of APP transcription [24]. These lines of evidence thus further support the interpretation that ApoE may normally serve a neuroprotective role in axon/synapse competition in the brain. Surprisingly, of the different ApoE alleles, ApoE4 appears to have the highest potency in stimulating APP transcription and Aβ monomer secretion in neurons [21, 24]. It has been suggested that the chronic effects of ApoE4 on APP and Aβ may be linked to the increased risk associated with the ApoE4 allele [24].

Unlike bacterial cell competition, eukaryotic cell competition, including axon/synapse competition, involves not only cellular components that directly compete with one another but also third-party glial cells that actively regulate this process [1]. These glial cells, such as microglia and astrocytes, not only express cytokines that control the timing and other aspects of axon/synapse competition, but also perform phagocytic functions such as dendritic spine pruning and Aβ clearance that control the execution and conclusion of this process. As such, controlling the activity of glial cells is likely also a key node in the regulation of axon/synapse competition. Indeed, a large body of evidence indicates that Aβ oligomers and monomers appear to not only act directly on competing neuronal structures but also engage and modulate the activity of glial cells during axon/synapse competition [1]. Importantly, recent studies show that, besides Aβ, ApoE also strongly impacts the activity of microglia and astrocytes and may also serve a neuroprotective role in axon/synapse competition through this indirect route [95100] (reviewed in [68, 69]).

A prominent mechanism by which microglia and astrocytes promote axon/synapse competition in the nervous system is through regulating inflammatory cytokines [1]. For example, in the retinogeniculate system, astrocyte-derived cytokine IL-33 has been found to be essential for neural activity dependent elimination of excess synapses [101]. At the vertebrate neuromuscular junction (NMJ), a classic model of activity-dependent axon competition, TNFα is also essential for activity-dependent elimination of supernumerary motor axon terminals [102]. In both the retinocollicular system and the somatosensory cortex, TNFRSF21 (or DR6), a TNFα receptor superfamily member, similarly promotes activity-dependent axon competition [103]. As mentioned, several studies suggest that, besides acting on neurons, ApoE may also protect competing axons/synapses by inhibiting inflammatory cytokine production in glial cells and as such dampen competition [95100]. Early studies show that ApoE strongly inhibits inflammatory activation of microglia in vitro [95]. Recent studies also show that ApoE possesses potent anti-inflammatory activity against macrophages, downregulating macrophage expression of pro-inflammatory genes while upregulating anti-inflammatory genes [96]. Furthermore, ApoE may also dampen glial cell inflammatory activity through well-established mechanisms such as promoting the efflux of cholesterol and other lipid molecules (reviewed in [1]). Consistent with this interpretation, deficiency in ApoE has been found to result in increased production of proinflammatory cytokines in vivo [97]. Deficiency in the ApoE receptors VLDLR or ApoER2 similarly results in enhanced proinflammatory phenotype in macrophages [96, 98]. Like Aβ, the anti-inflammatory effects of ApoE also appears to be concentration dependent, with high concentrations of ApoE promoting rather than suppressing inflammatory response in macrophages [99, 100]. Besides regulating glial expression of cytokines during axon/synapse competition, ApoE may also protect competing axons/synapses by regulating a second function of glial cells in this process, phagocytosis. For example, converging studies suggest that Aβ oligomers may directly promote glial phagocytosis and the elimination of competitor axons/synapses during competition [1]. Indeed, large numbers of studies show that ApoE promotes Aβ degradation by glial cells and may thus be protective to competing axons/synapses [39, 104109]. ApoE has been found to enhance Aβ trafficking and degradation by neprilysin in microglia through the modulation of microglial cholesterol levels [104, 105]. There is also evidence that ApoE may also promote Aβ degradation by astrocytes [106] (but see also [7]). In accordance with this interpretation, several groups have shown that ApoE is a strong ligand for the microglia specific Trem2 receptor [39, 107109], the activation of which has been shown by several groups to both promote microglial phagocytosis and inhibit inflammation [39, 107, 108, 110112]. Thus, these results support the interpretation that, like Aβ, low concentrations of ApoE may also serve a neuroprotective role in axon/synapse competition through regulating glial cell activity. Altogether, the experimental results discussed above suggest that ApoE may play a neuroprotective role in axon/synapse competition through a multitude of mechanisms including impeding toxic Aβ species formation, promoting Aβ clearance, and suppressing inflammatory cytokine production, and that the perturbation of this role of ApoE may contribute to the development of Alzheimer’s disease. Indeed, the ApoE4 allele has been found to be associated with high levels of soluble Aβ oligomers and microglial inflammatory activation in the human brain [12, 113115] as well as perturbed activity and immune regulation of human microglia and astrocytes [116122]. This suggests that compromises in the normal function of ApoE as discussed may play a role in the high risk associated with the ApoE4 allele.

Regulation of DNA damage repair by APP/AICD and its role in Alzheimer’s disease

Neural activity promotes Hebbian as well as non-Hebbian synaptic plasticity in the brain, both of which involve production of reactive oxygen species (ROS) [123130], a well-known inducer of DNA damage. Neural activity also induces the transcription of plasticity related genes, the activation of which requires double strand DNA break at gene regulatory regions [131134]. Thus, neural activity, while promoting plasticity, also induces DNA breaks or damages that are detrimental to the nervous system. In this regard, it is interesting to note that studies have long shown that APP, the precursor protein for Aβ, has an at times puzzling function in DNA damage repair [5567]. For example, studies have shown that large numbers of genotoxic agents induce the cleavage of the APP protein, leading to the release of a C-terminal proteolytic product (AICD) and an associated FE65 protein [55, 56], in a process dependent on the gamma-secretase complex [55, 135]. APP cleavage releasing AICD in turn promotes the nuclear translocation of FE65 [5760], which in the nucleus, binds to a Tip60-TRRAP histone acetyltransferase (HAT) complex, a complex that mediates histone H4 acetylation and DNA repair protein loading at DNA damage sites and plays an evolutionarily conserved role in DNA damage repair from yeast to mammals [6163]. FE65 also binds to and stabilizes the Bloom syndrome protein (BLM) [64], a DNA helicase with double strand break resection activity that promotes DNA repair by homologous recombination, especially at actively transcribed genes [65, 66]. Furthermore, the AICD/FE65 complex has also been found to regulate gene expression and decrease mitochondrial superoxide production [67]. Thus, these findings suggest that besides producing Aβ peptides that may directly mediate homeostatic and competitive plasticity, APP may potentially also participate in this process by producing an intracellular proteolytic product that promotes DNA damage repair and thus plays a potential protective role during synapse competition (Fig. 2). This may help reconcile the functional implication of APP in regulating DNA damage repair.

Figure 2. APP/AICD may be protective during axon/synapse competition.

Figure 2.

By producing an intracellular cleavage product (AICD), APP may potentially promote nuclear as well as potentially mitochondrial DNA repair through the adaptor protein FE65 and the TIP60/TRRAP histone acetylase complex. Figure created with BioRender.com.

Studies show that DNA damage in interphase cells and post-mitotic neurons leads to increased dynamics of both cytoplasmic and nuclear microtubules that promotes DNA repair [136138]. Endogenous tau proteins have also been found to directly bind to and protect neuronal DNA [139], while tau knockdown exacerbates double strand DNA breaks [140]. These findings thus raise a possibility that the nuclear phospho-tau that is prominently observed in brain neurons in tauopathies such as Alzheimer’s disease [141145] may be linked to the regulation of DNA damage repair [146148], providing a potential mechanistic explanation for the appearance of nuclear phospho-tau in various tauopathies. Importantly, besides in the nucleus [149152], pro-inflammatory cytokines, which as mentioned play key roles in axon/synapse pruning, have been found to also induce extensive DNA damage in the mitochondria [151, 153156]. Aβ oligomers, the proposed activity-dependent agent mediating axon/synapse elimination during competition, also directly target a significant number of mitochondrial regulators, many of which are linked to the production of reactive oxygen species [157162]. In addition, many components of the molecular machinery known for their function in nuclear DNA repair also localize to the mitochondria and perform key roles in maintaining the integrity of the mitochondrial genome [163166]. Thus, these findings suggest that, besides nuclear DNA repair, APP/AICD may also play a role in mitochondrial DNA repair, a process that may potentially be more relevant to activity-dependent axon/synapse competition, since mitochondrial DNA may be the main target of the reactive oxygen species produced in this process. Indeed, studies have reported the localization of APP cleavage pathway components and products in the mitochondria. However, the interpretation of these results has been complicated by factors including the frequent use of gene overexpression in these experiments (reviewed in [167]). Nonetheless, recent studies show that mitochondrial DNA damage and release into the cytoplasm and the extracellular environment is closely associated with aging and the development of neuroinflammation and cognitive function decline in humans [168171]. Alzheimer’s disease fibroblasts have also been found to show increased levels of cytoplasmic mitochondrial DNA [172]. This suggests that both nuclear and mitochondrial DNA damage may play a role in the development of Alzheimer’s disease and may be potential targets for clinical intervention [173, 174].

Relationship between roles played by oligomeric Aβ and ApoE in physiological synapse elimination and in Alzheimer’s disease.

In axon/synapse competition, competing axons/synapses must not only protect themselves but also eliminate competitors. Strong evidence indicates that neurotoxic Aβ oligomers induced by high levels of neural circuit activity play a large role in effecting competitor elimination in this process [1]. Among the evidence, it has been found that PirB, a high affinity Aβ oligomer receptor, and related signaling pathway play a crucial role in ocular dominance plasticity and mutations in this pathway severely disrupt cortical synapse reorganization induced by monocular deprivation [175178]. Besides PirB, recent studies have also identified several other high affinity Aβ oligomer receptors and show that they play similar roles in homeostatic and competitive plasticity across the nervous system [179198]. Furthermore, besides specific receptors, studies show that Aβ oligomers also mediate competitor elimination through general mechanisms that target unique membrane lipid changes induced by synapse competition [2531, 3436, 3841]. In addition, unlike the protective role played by astrocytic ApoE, recent studies show that neuronal ApoE may instead promote competitor elimination during axon/synapse competition [199214]. In this section, I will discuss these new findings and highlight how they may shed new light on the widespread loss of synapses as well as the selective vulnerability to neurodegeneration of ApoE expressing neurons in Alzheimer’s disease.

Aβ oligomer receptors and the targeting of different classes of dendritic spines during synaptic plasticity and in Alzheimer’s disease

Dendritic spines are the most common postsynaptic component of excitatory synapses in the brain and are targets for activity-dependent competitive elimination during axon/synapse competition. Spine elimination is typically preceded by synaptic depression, which involves at least two different mechanisms, metabotropic glutamate receptor (mGluR)- and NMDA-type glutamate receptor (NMDAR)-dependent long-term depression (LTD) [215, 216]. Spines come in different shapes and sizes. Large spines, frequently of a mushroom shape, typically contain stacks of endoplasmic reticulum (ER) known as the spine apparatus, while small spines lack the spine apparatus [217221]. Interestingly, recent studies show that only large spines appear to be capable of undergoing mGluR-dependent LTD [222, 223] and require the activation of both mGluR- and NMDAR-dependent LTD to undergo size shrinkage [224] (but see also [189]), while small spines can undergo size shrinkage upon induction of NMDAR-dependent LTD alone.

Importantly, independent studies show that different high affinity Aβ oligomer receptors appear to differentially regulate mGluR- and NMDAR-dependent signaling and LTD at excitatory synapses [179198]. This raises a possibility that Aβ oligomers may target and regulate the function of different types of dendritic spines through different combinations of Aβ oligomer receptors (Fig. 3). For example, Aβ oligomers have been found to bind with high affinity to the cellular prion protein (Prpc) [179], which is highly enriched at excitatory postsynaptic sites and forms a complex with mGluR5. Upon Aβ oligomer binding, cellular prion protein traps mGluR5 in the plasma membrane, leading to spine loss through activation of the nonreceptor tyrosine kinase Fyn [180182]. In vivo, increased interactions between mGluR5 and Fyn have been observed in the barrel cortex following whisker trimming, a well-known model of competitive plasticity [183], suggesting that this regulatory mechanism takes place under physiological conditions. Thus, the cellular prion protein may mediate Aβ oligomer effects on a select group of dendritic spines (potentially large spines since only they appear capable of undergoing mGluR-LTD) through activating mGluR-LTD or related pathways [184, 185]. On the other hand, while the direct binding of Aβ oligomers to NMDARs has not been observed, Aβ oligomers have been found to bind to several NMDAR associated surface receptors at excitatory synapses. Aβ oligomers bind with high affinity to EphB2 [186], a tyrosine kinase receptor that plays key roles in the synaptic localization and activity of NMDARs [225228]. Aβ oligomers also bind to and affect the function of several planar cell polarity (PCP) proteins such as Frizzled and Celsr3 [187, 188] that play similarly critical roles in the formation and function of excitatory synapses including the NMDARs [229232]. Furthermore, Aβ oligomers also inhibit NMDAR-dependent long-term potentiation (LTP) through the cellular prion protein and mGluR5 [185]. Most important, while the potential effects of spine size, if any, remain unclear, large numbers of studies consistently show that NMDAR activity is required for both Aβ oligomer-induced synaptic depression and Aβ oligomer-induced shrinkage at spines, possibly through both its ionotropic and metabotropic activities [189198]. These results therefore support the possibility that during synapse competition, Aβ oligomers may potentially target and eliminate large spines through both mGluR-linked (e.g., the cellular prion protein) and NMDAR-linked receptors but target small spines through NMDAR-linked receptors alone. This not only provides further evidence supportive of the role of Aβ in synaptic competition but is also consistent with observations that the prion protein forms a complex with Aβ and mediates the toxicity of soluble Aβ aggregates in the Alzheimer’s disease brain [233235] and the levels of NMDARs are severely perturbed [236239].

Figure 3. Mechanisms by which Aβ oligomers may potentially target different classes of dendritic spines.

Figure 3.

Studies (see text) suggest that only large but not small spines can undergo and require mGluR-dependent LTD for shrinkage, while both large and small spines require NMDAR-dependent LTD for shrinkage. Thus, a potential possibility is that Aβ oligomers may target and induce the shrinkage of different classes of spines through activating different combinations of Aβ oligomer receptors at these spines. Figure created with BioRender.com.

Aβ interaction with phosphatidylserine and the targeting of axons/synapses under physiological and diseased conditions

Phosphatidylserine (PS), a phospholipid that normally resides in the inner leaflet of the plasma membrane but is exposed on the outer leaflet upon induction of apoptosis, is a well-known “eat-me” signal that promotes cell debris clearance during programmed cell death [240, 241]. Recent studies show that this signal is conserved and plays a critical role in activity-dependent axon/synapse competition [242245]. Notably, phosphatidylserine exposure on pre- and post-synaptic membranes has been found to be developmentally regulated and coincide with the period of activity-dependent synapse pruning in vivo, and blockade of its interaction with phosphatidylserine receptors on microglia by multiple approaches all results in compromised microglial engulfment and elimination of supernumerary synapses during normal development [242245]. Aβ oligomers have potent membrane disrupting activities mediated by mechanisms including pore formation [246248], lipid extraction [249, 250], and partial insertion as well as carpet-like mechanisms [251254]. These mechanisms may in part mediate the pruning of weak axons/synapses in activity-dependent competition [1]. As such, the timing and the kinetics of Aβ oligomer formation are likely key factors in axon/synapse competition. Indeed, studies show that Aβ oligomer formation is regulated by many factors including a complex interaction with plasma membrane components [2530]. For example, the different phospholipids of the plasma membrane that reside in the inner or outer leaflet appear to impact Aβ oligomerization very differently [25, 26]. Phosphatidylserine-like lipids that are only exposed in the outer leaflet upon induction of processes such as apoptosis accelerate, while phosphatidylcholine-like lipids that normally populate the outer leaflet slow down the aggregation of Aβ peptide fragments [25, 26]. Likely as a result, the extent of cell surface exposure of phosphatidylserine has been found to determine the susceptibility of cells to Aβ toxicity while phosphatidylcholine, which is normally found in the outer leaflet, renders protection for these cells [2729]. Recent studies have further shown that when incorporated into the outer leaflet of a lipid bilayer, phosphatidylserine, but not phosphatidylcholine, can trigger rapid Aβ oligomerization even at subnanomolar concentrations and as a result induce extensive membrane pore formation [30]. These unique effects of phosphatidylserine suggest a potential self-amplifying mechanism during axon/synapse competition in which phosphatidylserine-induced Aβ oligomerization may possibly act to increase the strength or duration of the initial “eat-me” signal produced by phosphatidylserine exposure and as such facilitate the specific tagging and elimination of weak axons/synapses (Fig. 4).

Figure 4. Mechanisms by which phosphatidylserine (PS) may promote Aβ oligomerization, amplify the “eat-me” signal, and tag synapses for phagocytosis.

Figure 4.

PS on the outer leaflet of the plasma membrane, exposed initially by competitive interaction between synapses, may potentially catalyze the local oligomerization of Aβ at the tagged specific synapse and thus amplify the initial signal. PS and Aβ oligomers have both been shown to directly and indirectly activate several microglial receptors that promote phagocytosis and thus likely synapse pruning. Together, it appears as if they act in a concerted manner to promote synapse pruning. Figure created with BioRender.com.

In support of above interpretation, Aβ oligomers have been found to enhance the surface exposure of phosphatidylserine and phosphatidylserine-directed engulfment of spine synapses [31], suggesting a mutually reinforcing interaction between phosphatidylserine and Aβ oligomers. Several receptors known to bind to phosphatidylserine and promote cell debris phagocytosis and clearance have been found to also bind to and be activated by Aβ oligomers [3241]. For example, the complement cascade and receptors are well known to play a prominent role in activity-dependent axon/synapse competition [255257], by recognizing as well as being activated by phosphatidylserine in this process [32, 33]. Importantly, large numbers of studies show that Aβ oligomers also directly and independently activate both the classical and the alternative complement pathway [3436]. Thus, phosphatidylserine and Aβ oligomers may act in a concerted manner in the activation of the complement-dependent clearance pathways. Similarly, the TAM receptor tyrosine kinases Tyro3, Axl, and Mer, which play key roles in synapse competition in the retinogeniculate system [258], are also known to be activated by phosphatidylserine through bridging proteins [37]. Recent studies have shown that microglia may also employ the TAM receptors to detect and engulf Aβ aggregates associated with amyloid plaques [38]. Furthermore, the microglia specific Trem2 receptor, the activation of which promotes microglial phagocytosis [39, 107, 108, 111, 112], is known to bind with significant affinity and be activated by phosphatidylserine [39, 40]. Independent studies show that it can also bind directly to Aβ oligomers with nanomolar affinity and this interaction is required for both Aβ regulation of downstream signaling activity and Aβ degradation by microglia [41]. Altogether, these results strongly suggest that, besides activation of specific surface receptors, the regulation of Aβ oligomer formation by membrane-exposed phosphatidylserine may also promote activity-dependent axon/synapse competition by amplifying the “eat-me” signal and enhancing the specific tagging and elimination of weak axons/synapses. Importantly, large numbers of studies show that the dysregulation of this mechanism likely plays a significant role in Alzheimer’s disease [259265]. For example, studies show that caspase 3, a key effector caspase in apoptosis, inactivates flippases such as ATP11A and ATP11C and activates scramblases such as Xkr8, both of which promote the cell surface exposure of phosphatidylserine [259261]. In the Alzheimer’s disease brain, the levels of active caspase 3 have been found to be elevated especially in the postsynaptic density [262265], potentially contributing to the extensive loss of synapses.

Regulation of MHCI and related competition genes by neuronal ApoE and the selective vulnerability of ApoE expressing neurons in Alzheimer’s disease

In previous sections, I discuss evidence indicating that astrocytic ApoE acts as a neuroprotective agent in axon/synapse competition by, among others, upregulating APP gene transcription and directly and indirectly suppressing the assembly of toxic Aβ species. However, new studies indicate that this is unlikely the sole function of ApoE. Indeed, ApoE, especially ApoE proteins of neuronal origin, may be part of a regulatory network that normally promotes the expression and the activity of a specific set of genes directly involved in axon/synapse competition [199214] (Fig. 5). ApoE is most highly expressed in astrocytes in the normal brain. However, it is also expressed in a select group of neurons in a manner dependent on neural activity [73, 74]. The level of ApoE expression in these neurons not only strongly correlates, on a cell-by-cell basis, with that of immune response genes in these same neurons but also regulates the expression of these immune response genes [199]. Many of these ApoE-regulated genes in turn play key roles in axon/synapse competition. For example, neuronal ApoE regulates the expression of MHCI, the class I major histocompatibility complex and a molecular partner of PirB that play pivotal roles in ocular dominance plasticity [177, 178, 200, 201]. Neuronal ApoE also regulates the expression of several other genes such as Tap2 that interact and cooperate with MHCI at the molecular level [202]. It also regulates the expression of C1qa, a component of the complement cascade with widespread roles in activity-dependent synapse elimination [255, 256]. Moreover, it regulates the neuronal expression of interleukin 4, a cytokine that potently modulates brain microglial pathways and neuronal network activity [203, 204]. Thus, neuronal ApoE appears to play a pivotal role in coordinating the expression of genes involved in axon/synapse competition and as such promoting competitive plasticity in the normal brain.

Figure 5. Autocrine and paracrine ApoE may together regulate a gene network that promotes axon/synapse competition.

Figure 5.

Studies (see text for detailed discussion) indicate that neuronal ApoE promotes, in an autocrine manner, the expression in the same neurons of large numbers of genes, including MHCI, Tap2, and C1qa, that directly participate in axon/synapse competition. Astrocyte-derived ApoE has also been shown to increase APP transcription and Aβ secretion by neurons. In addition, as shown in Fig. 1, ApoE also appears to directly interact with Aβ. Thus, ApoE appears to be a key part of the gene network that regulates axon/synapse competition. Figure created with BioRender.com.

Importantly, the regulation by ApoE of this group of immune response genes with demonstrated or likely roles in axon/synapse competition appears to be linked to the selective vulnerability of these expressing neurons in Alzheimer’s disease. For example, functional reduction of MHCI has been found to ameliorate tau pathology in both ApoE4-expressing primary neurons and pathological tau-expressing mouse hippocampi [199]. Interestingly, previous studies have shown that Stat1, a transcriptional factor that mediates JAK2 driven elimination of inactive synapses during activity-dependent competition [205], binds to the enhancer and regulates the transcription of ApoE gene [206]. Similarly, the neuronal expression of RORβ, another marker for selectively vulnerable neurons in Alzheimer’s disease [207], is not only required for the normal formation of the barrel cortex in a well-known activity-dependent competitive process, and sufficient to induce barrel-like neuronal clusters when overexpressed [208, 209], but it is also regulated by ApoE4 [210]. Furthermore, Stat1/2 also bind to the promoter and promote the expression of RORβ gene [211]. Conversely, strong Stat1 activity has been linked to suppressed expression of MEF2C, a key transcriptional factor that confers resilience to neurodegeneration in Alzheimer’s disease and protect neurons when overexpressed [212214]. Thus, together with the results showing the regulation of APP transcription and Aβ secretion by ApoE, these findings suggest that Aβ, ApoE, MHCI, Stat1, RORβ, and other genes may normally form a regulatory network that promotes neural activity-dependent axon/synapse competition under physiological conditions. Perturbation of the activity of this network may upset the delicate balance in its neuroprotective and neurotoxic activities and lead to the development of Alzheimer’s disease. This may in part explain the selective vulnerability of ApoE and RORβ expressing neurons in Alzheimer’s disease since the expression of these genes in these neurons suggest that they may normally be engaged in competitive plasticity under physiological conditions and thus especially vulnerable to perturbations in the process.

Relationship between Aβ, homeostatic and competitive plasticity, and sleep

An intriguing observation of competitive synaptic plasticity was made over twenty years ago when it was found that sleep robustly enhances the effects of competitive plasticity such as those observed following monocular deprivation [266]. Studies since have shown that rapid eye movement (REM) sleep, a sleep stage when dendritic spine elimination is especially prevalent in the brain [267, 268], appears to play an important role in ocular dominance plasticity and related processes [269272]. Aβ levels in the brain are regulated by the sleep-wake cycle, being dramatically up-regulated upon sleep deprivation [42]. As mentioned, a large body of evidence also suggests that Aβ oligomers that form in the brain as a result of high-level neural activity may directly mediate activity-dependent axon/synapse competition [1]. These findings thus raise the question of whether there are mechanistic links between Aβ, sleep, and activity-dependent synapse competition. Recent studies show that this may indeed be the case, potentially explaining the well-known role of sleep perturbation in Alzheimer’s disease [4254].

First, strong evidence indicates that Aβ oligomers may directly induce sleep [4350]. Aβ levels in the human brain are generally low in the morning, but gradually increase during the day before peaking at night [42]. Aβ oligomerization has been shown to be likely regulated by a concentration-dependent nucleation mechanism [273, 274]. As the day goes on and the levels of Aβ in the brain rise, the levels of Aβ oligomers in the brain are likely to also rise. As mentioned, one of the high-affinity Aβ oligomer receptors in the brain is the cellular prion protein, which activates mGluR5 and Fyn and induces synaptic depression and spine loss upon Aβ oligomer binding [179182]. Interestingly, recent studies show that in both zebrafish and mice, Aβ oligomers also act through the cellular prion protein to regulate sleep [43, 44]. Exogenous application of long Aβ oligomers, for example, has been found to dampen brain neuronal activity, increase sleep, and reduce waking activity in zebrafish, an effect prevented by prion gene mutation [43]. The increased sleep is linked to increases in the number of sleep bouts but not the lengthening of individual bouts, indicating that long Aβ oligomers boost sleep initiation. Conversely, inhibiting the activities of mGluR5 and Fyn both blocks the sleep-inducing effects of long Aβ oligomers [43]. Prior to this, studies have also shown that loss-of-function mutations in the cellular prion gene result in altered circadian activity rhythms and sleep in mice [45, 46]. Knockout of mGluR5 similarly results in severe dysregulation of sleep-wake homeostasis, including a lack of recovery sleep after sleep deprivation [47]. Furthermore, in humans, increased levels of mGluR5 after sleep loss have been found to tightly correlate with biomarkers of elevated sleep need [48]. Thus, these results all suggest that there are strong molecular links between Aβ and sleep. Indeed, independent studies show that the function of cellular prion proteins is required for the nighttime increases in melatonin [49], a sleep-promoting hormone that plays key roles in sleep cycle regulation [275, 276]. Evidence also suggests that activity of the cellular prion protein may increase the degradation of norepinephrine [50], a hormone that normally promotes wakefulness [277279]. Activity of the glymphatic system normally increases during sleep and promotes the clearance of metabolic waste from the brain [280, 281]. Interestingly, signaling by mGluR5 as well as by adenosine, the latter being a critical sleep-promoting metabolite [282284], have both been found to promote astrocyte expression of Aquaporin 4 [285, 286], a water channel with crucial roles in the function of the glymphatic system [287, 288]. Thus, these findings indicate that Aβ, a candidate molecular agent for mediating competitive synaptic plasticity, may potentially also directly regulate sleep, a brain state with crucial roles in competitive plasticity.

Besides sleep induction, research on the mechanisms of synaptic scaling-down, a form of homeostatic plasticity, during sleep have also revealed molecular links between Aβ oligomers and synaptic changes in sleep and provided further support for a role of Aβ and sleep in homeostatic and competitive synaptic plasticity [5153] (Fig. 6). Sleep plays important roles in many aspects of brain function [289, 290]. Among the hypotheses proposed on the function of sleep, the synaptic homeostasis hypothesis (SHY) proposes that a key function of sleep is to restore synaptic homeostasis in the brain after a net increase in synaptic strength following a day of wakefulness [291]. Indeed, three-dimensional electron microscopy (EM) studies show that the average axon-spine interface (ASI) in the mouse motor and sensory cortices decreases by ~18% after sleep and the decrease is proportional to ASI size, indicating synaptic scaling [292294](but see also [295]). The scaling appears to be selective, sparing synapses that are large and lack recycling endosomes (spines with and without a spine apparatus, however, both appear to undergo scaling) [292]. Interestingly, independent studies show that mGluR5, the molecular partner of the cellular prion protein that mediates the induction of sleep by Aβ oligomers [43, 44, 47, 48], also plays a crucial role in synaptic scaling-down during sleep [51, 52]. mGluR5 and related mGluR1 have been found to trigger synaptic scaling-down in sleep by activating ligand-independent signaling through a mechanism dependent on Homer1a, a postsynaptic cytoskeletal protein induced by sleep loss [53]. Specifically, these studies find that Homer1a levels in the postsynaptic density are increased by the sleep-promoting adenosine but decreased by the wake-promoting norepinephrine [51]. As a result, during sleep, high levels of Homer1a accumulate at the postsynaptic density and, through altering postsynaptic organization, activate mGluR1/5 signaling, which then promotes the removal of surface AMPA receptors and the weakening of these synapses [51]. Indeed, mutations in Homer1a and inhibition of mGluR1/5 activity have both been found to block sleep cycle-associated changes in synaptic AMPA receptors [51]. These results thus implicate a critical role played by Homer1a-activated mGluR1/5 signaling in synaptic scaling-down during sleep. Importantly, both Homer1a and mGluR5 have also been found to be required for the establishment of the contralateral bias as well as the maintenance of ocular dominance in the mouse cortex [54]. This supports the interpretation that sleep-associated synaptic scaling-down may be physiologically linked to and play a key role in competitive plasticity, a process also associated with and regulated by sleep [269272].

Figure 6. Mechanisms by which Aβ oligomers may potentially regulate the synaptic specificity of Homer1a targeting during homeostatic synaptic scaling-down in sleep.

Figure 6.

Synaptic targeting of Homer1a has been shown to play a key role in homeostatic down-scaling in sleep. Significant evidence (see text) suggests that ERK cascade activity may regulate the synapse specificity of Homer1a targeting. As shown in Fig. 3, Aβ oligomers can activate both mGluR5 and NMDAR signaling, which have also both been shown to activate ERK. Thus, a potential determinant for the synapse specificity of Homer1a-medaietd down-scaling may be the local concentration of Aβ oligomers near a synapse and the degree and combination of Aβ oligomer receptors they may activate. Figure created with BioRender.com.

Furthermore, studies suggest that the regulation of synaptic scaling-down during sleep, especially the specificity of the synapses that are being targeted, may be linked to the activation of Aβ oligomer receptors at these synapses [180, 296306]. This potentially parallels the differential targeting of large and small spines by Aβ oligomers, through the different combination of receptors, as discussed in the previous section. For example, in parallel to the morphological scaling of synapses during sleep, synaptic targeting of Homer1a has also been found to be selective [296298], in a process dependent on the activation of the ERK serine/threonine kinase [296, 297, 299]. Interestingly, the neuronal ERK cascade has been found to be activated by cellular prion protein signaling through the Fyn receptor tyrosine kinase [300303], both of which are in turn activated by Aβ oligomer binding to the prion protein [180]. In addition, NMDARs, which are required for and mediate Aβ oligomer-induced synaptic depression and spine shrinkage at both large and small spines, have also been found to regulate the activity of the ERK cascade in neurons [304306]. Thus, these results suggest that, besides regulating sleep, Aβ oligomers may possibly also regulate the specificity of synaptic scaling-down during sleep through regulating ERK and Homer1a activity at specific synapses. As such, Aβ appears to play a central role in orchestrating and coordinating many aspects of synaptic reorganization and homeostatic and competitive plasticity during sleep. This may explain the prominent role that sleep disturbances play in the development of Alzheimer’s disease.

Concluding remarks

Aβ and ApoE are two of the biggest players in Alzheimer’s disease pathology, but they also play physiological roles in the healthy brain in processes including activity-dependent homeostatic and competitive plasticity. In this review, I have summarized a large body of research showing that the roles played by Aβ and ApoE in Alzheimer’s disease can be unified with those they play in physiological processes and that it is the perturbation of these physiological processes that underlies the development of Alzheimer’s disease. For example, being part of a normal mechanism that regulates activity-dependent homeostatic and competitive plasticity in the brain, the formation of Aβ oligomers likely, under physiological conditions, promotes the removal of supernumerary synapses, through both activating different combinations of receptors at different dendritic spines and amplifying general cell surface “eat-me” signals. In Alzheimer’s disease, however, perturbation of these processes leads to excess Aβ oligomer formation. This may not only provoke an intense neuroinflammatory response but also lead to extensive dendritic spine pruning and loss, both common pathologies observed in the Alzheimer’s disease brain. Similarly, the ApoE protein likely normally regulates the kinetics of Aβ aggregation and turnover as well as glial lipid metabolism (astrocytic ApoE) as well as promotes the expression of genes involved in the physiological process of competitive synaptic plasticity, by forming a gene regulatory network with Aβ and other molecules (neuronal ApoE). In Alzheimer’s disease, however, perturbation of these ApoE regulated processes not only contributes to excess Aβ oligomer formation but also underlies the selective vulnerability of ApoE expressing neurons in the diseased brain. Furthermore, Aβ oligomers not only induce sleep but may also orchestrate the processes of synaptic scaling-down and spine pruning, plasticity processes linked to this brain state. Thus, this body of research in aggregate provides compelling evidence that Alzheimer’s disease is a disease of competitive synaptic plasticity gone wrong, a perspective that may promote advance in this area.

Funding

Funding was provided in part by NIH/NCATS through CTSA award UL1TR002373 to the UW Institute for Clinical and Translational Research.

Footnotes

Conflict of Interest

The author has no conflict of interest to report.

This manuscript has been accepted by The Journal of Alzheimer’s Disease (JAD, https://www.j-alz.com/)

References:

  • [1].Huang Z (2023) A Function of Amyloid-beta in Mediating Activity-Dependent Axon/Synapse Competition May Unify Its Roles in Brain Physiology and Pathology. J Alzheimers Dis 92, 29–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [2].Strittmatter WJ, Weisgraber KH, Huang DY, Dong LM, Salvesen GS, Pericak-Vance M, Schmechel D, Saunders AM, Goldgaber D, Roses AD (1993) Binding of human apolipoprotein E to synthetic amyloid beta peptide: isoform-specific effects and implications for late-onset Alzheimer disease. Proc Natl Acad Sci U S A 90, 8098–8102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [3].Aleshkov S, Abraham CR, Zannis VI (1997) Interaction of nascent ApoE2, ApoE3, and ApoE4 isoforms expressed in mammalian cells with amyloid peptide beta (1-40). Relevance to Alzheimer’s disease. Biochemistry 36, 10571–10580. [DOI] [PubMed] [Google Scholar]
  • [4].LaDu MJ, Pederson TM, Frail DE, Reardon CA, Getz GS, Falduto MT (1995) Purification of apolipoprotein E attenuates isoform-specific binding to beta-amyloid. J Biol Chem 270, 9039–9042. [DOI] [PubMed] [Google Scholar]
  • [5].Tokuda T, Calero M, Matsubara E, Vidal R, Kumar A, Permanne B, Zlokovic B, Smith JD, Ladu MJ, Rostagno A, Frangione B, Ghiso J (2000) Lipidation of apolipoprotein E influences its isoform-specific interaction with Alzheimer’s amyloid beta peptides. Biochem J 348 Pt 2, 359–365. [PMC free article] [PubMed] [Google Scholar]
  • [6].Sanan DA, Weisgraber KH, Russell SJ, Mahley RW, Huang D, Saunders A, Schmechel D, Wisniewski T, Frangione B, Roses AD, et al. (1994) Apolipoprotein E associates with beta amyloid peptide of Alzheimer’s disease to form novel monofibrils. Isoform apoE4 associates more efficiently than apoE3. J Clin Invest 94, 860–869. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [7].Verghese PB, Castellano JM, Garai K, Wang Y, Jiang H, Shah A, Bu G, Frieden C, Holtzman DM (2013) ApoE influences amyloid-beta (Abeta) clearance despite minimal apoE/Abeta association in physiological conditions. Proc Natl Acad Sci U S A 110, E1807–1816. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [8].Garai K, Verghese PB, Baban B, Holtzman DM, Frieden C (2014) The binding of apolipoprotein E to oligomers and fibrils of amyloid-beta alters the kinetics of amyloid aggregation. Biochemistry 53, 6323–6331. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [9].Ly S, Altman R, Petrlova J, Lin Y, Hilt S, Huser T, Laurence TA, Voss JC (2013) Binding of apolipoprotein E inhibits the oligomer growth of amyloid-beta peptide in solution as determined by fluorescence cross-correlation spectroscopy. J Biol Chem 288, 11628–11635. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [10].Ghosh S, Sil TB, Dolai S, Garai K (2019) High-affinity multivalent interactions between apolipoprotein E and the oligomers of amyloid-beta. FEBS J 286, 4737–4753. [DOI] [PubMed] [Google Scholar]
  • [11].Fitz NF, Nam KN, Wolfe CM, Letronne F, Playso BE, Iordanova BE, Kozai TDY, Biedrzycki RJ, Kagan VE, Tyurina YY, Han X, Lefterov I, Koldamova R (2021) Phospholipids of APOE lipoproteins activate microglia in an isoform-specific manner in preclinical models of Alzheimer’s disease. Nat Commun 12, 3416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [12].Hashimoto T, Serrano-Pozo A, Hori Y, Adams KW, Takeda S, Banerji AO, Mitani A, Joyner D, Thyssen DH, Bacskai BJ, Frosch MP, Spires-Jones TL, Finn MB, Holtzman DM, Hyman BT (2012) Apolipoprotein E, especially apolipoprotein E4, increases the oligomerization of amyloid beta peptide. J Neurosci 32, 15181–15192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [13].Hashimoto T, Adams KW, Fan Z, McLean PJ, Hyman BT (2011) Characterization of oligomer formation of amyloid-beta peptide using a split-luciferase complementation assay. J Biol Chem 286, 27081–27091. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [14].Ma J, Yee A, Brewer HB Jr., Das S, Potter H (1994) Amyloid-associated proteins alpha 1-antichymotrypsin and apolipoprotein E promote assembly of Alzheimer beta-protein into filaments. Nature 372, 92–94. [DOI] [PubMed] [Google Scholar]
  • [15].Yang T, Li S, Xu H, Walsh DM, Selkoe DJ (2017) Large Soluble Oligomers of Amyloid beta-Protein from Alzheimer Brain Are Far Less Neuroactive Than the Smaller Oligomers to Which They Dissociate. J Neurosci 37, 152–163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [16].Bales KR, Verina T, Cummins DJ, Du Y, Dodel RC, Saura J, Fishman CE, DeLong CA, Piccardo P, Petegnief V, Ghetti B, Paul SM (1999) Apolipoprotein E is essential for amyloid deposition in the APP(V717F) transgenic mouse model of Alzheimer’s disease. Proc Natl Acad Sci U S A 96, 15233–15238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [17].Huynh TV, Liao F, Francis CM, Robinson GO, Serrano JR, Jiang H, Roh J, Finn MB, Sullivan PM, Esparza TJ, Stewart FR, Mahan TE, Ulrich JD, Cole T, Holtzman DM (2017) Age-Dependent Effects of apoE Reduction Using Antisense Oligonucleotides in a Model of beta-amyloidosis. Neuron 96, 1013–1023 e1014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [18].Liu CC, Zhao N, Fu Y, Wang N, Linares C, Tsai CW, Bu G (2017) ApoE4 Accelerates Early Seeding of Amyloid Pathology. Neuron 96, 1024–1032 e1023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [19].Ulrich JD, Ulland TK, Mahan TE, Nystrom S, Nilsson KP, Song WM, Zhou Y, Reinartz M, Choi S, Jiang H, Stewart FR, Anderson E, Wang Y, Colonna M, Holtzman DM (2018) ApoE facilitates the microglial response to amyloid plaque pathology. J Exp Med 215, 1047–1058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [20].Bien-Ly N, Gillespie AK, Walker D, Yoon SY, Huang Y (2012) Reducing human apolipoprotein E levels attenuates age-dependent Abeta accumulation in mutant human amyloid precursor protein transgenic mice. J Neurosci 32, 4803–4811. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [21].Huang YA, Zhou B, Wernig M, Sudhof TC (2017) ApoE2, ApoE3, and ApoE4 Differentially Stimulate APP Transcription and Abeta Secretion. Cell 168, 427–441 e421. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [22].Wang H, Kulas JA, Wang C, Holtzman DM, Ferris HA, Hansen SB (2021) Regulation of beta-amyloid production in neurons by astrocyte-derived cholesterol. Proc Natl Acad Sci U S A 118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [23].Barrett PJ, Song Y, Van Horn WD, Hustedt EJ, Schafer JM, Hadziselimovic A, Beel AJ, Sanders CR (2012) The amyloid precursor protein has a flexible transmembrane domain and binds cholesterol. Science 336, 1168–1171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [24].Huang YA, Zhou B, Nabet AM, Wernig M, Sudhof TC (2019) Differential Signaling Mediated by ApoE2, ApoE3, and ApoE4 in Human Neurons Parallels Alzheimer’s Disease Risk. J Neurosci 39, 7408–7427. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [25].Budvytyte R, Valincius G (2023) The interactions of amyloid beta aggregates with phospholipid membranes and the implications for neurodegeneration. Biochem Soc Trans 51, 147–159. [DOI] [PubMed] [Google Scholar]
  • [26].Nagarathinam A, Hoflinger P, Buhler A, Schafer C, McGovern G, Jeffrey M, Staufenbiel M, Jucker M, Baumann F (2013) Membrane-anchored Abeta accelerates amyloid formation and exacerbates amyloid-associated toxicity in mice. J Neurosci 33, 19284–19294. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [27].Ko M, Hattori T, Abdullah M, Gong JS, Yamane T, Michikawa M (2016) Phosphatidylcholine protects neurons from toxic effects of amyloid beta-protein in culture. Brain Res 1642, 376–383. [DOI] [PubMed] [Google Scholar]
  • [28].Del Mar Martinez-Senac M, Villalain J, Gomez-Fernandez JC (1999) Structure of the Alzheimer beta-amyloid peptide (25–35) and its interaction with negatively charged phospholipid vesicles. Eur J Biochem 265, 744–753. [DOI] [PubMed] [Google Scholar]
  • [29].Simakova O, Arispe NJ (2007) The cell-selective neurotoxicity of the Alzheimer’s Abeta peptide is determined by surface phosphatidylserine and cytosolic ATP levels. Membrane binding is required for Abeta toxicity. J Neurosci 27, 13719–13729. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [30].Robinson J, Sarangi NK, Keyes TE (2023) Role of phosphatidylserine in amyloid-beta oligomerization at asymmetric phospholipid bilayers. Phys Chem Chem Phys 25, 7648–7661. [DOI] [PubMed] [Google Scholar]
  • [31].Rueda-Carrasco J, Sokolova D, Lee SE, Childs T, Jurcakova N, Crowley G, De Schepper S, Ge JZ, Lachica JI, Toomey CE, Freeman OJ, Hardy J, Barnes SJ, Lashley T, Stevens B, Chang S, Hong S (2023) Microglia-synapse engulfment via PtdSer-TREM2 ameliorates neuronal hyperactivity in Alzheimer’s disease models. EMBO J 42, e113246. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [32].Paidassi H, Tacnet-Delorme P, Garlatti V, Darnault C, Ghebrehiwet B, Gaboriaud C, Arlaud GJ, Frachet P (2008) C1q binds phosphatidylserine and likely acts as a multiligand-bridging molecule in apoptotic cell recognition. J Immunol 180, 2329–2338. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [33].Mevorach D, Mascarenhas JO, Gershov D, Elkon KB (1998) Complement-dependent clearance of apoptotic cells by human macrophages. J Exp Med 188, 2313–2320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [34].Bradt BM, Kolb WP, Cooper NR (1998) Complement-dependent proinflammatory properties of the Alzheimer’s disease beta-peptide. J Exp Med 188, 431–438. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [35].Rogers J, Cooper NR, Webster S, Schultz J, McGeer PL, Styren SD, Civin WH, Brachova L, Bradt B, Ward P, et al. (1992) Complement activation by beta-amyloid in Alzheimer disease. Proc Natl Acad Sci U S A 89, 10016–10020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [36].Jiang H, Burdick D, Glabe CG, Cotman CW, Tenner AJ (1994) beta-Amyloid activates complement by binding to a specific region of the collagen-like domain of the C1q A chain. J Immunol 152, 5050–5059. [PubMed] [Google Scholar]
  • [37].Lew ED, Oh J, Burrola PG, Lax I, Zagorska A, Traves PG, Schlessinger J, Lemke G (2014) Differential TAM receptor-ligand-phospholipid interactions delimit differential TAM bioactivities. Elife 3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [38].Huang Y, Happonen KE, Burrola PG, O’Connor C, Hah N, Huang L, Nimmerjahn A, Lemke G (2021) Microglia use TAM receptors to detect and engulf amyloid beta plaques. Nat Immunol 22, 586–594. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [39].Wang Y, Cella M, Mallinson K, Ulrich JD, Young KL, Robinette ML, Gilfillan S, Krishnan GM, Sudhakar S, Zinselmeyer BH, Holtzman DM, Cirrito JR, Colonna M (2015) TREM2 lipid sensing sustains the microglial response in an Alzheimer’s disease model. Cell 160, 1061–1071. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [40].Shirotani K, Hori Y, Yoshizaki R, Higuchi E, Colonna M, Saito T, Hashimoto S, Saito T, Saido TC, Iwata N (2019) Aminophospholipids are signal-transducing TREM2 ligands on apoptotic cells. Sci Rep 9, 7508. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [41].Zhao Y, Wu X, Li X, Jiang LL, Gui X, Liu Y, Sun Y, Zhu B, Pina-Crespo JC, Zhang M, Zhang N, Chen X, Bu G, An Z, Huang TY, Xu H (2018) TREM2 Is a Receptor for beta-Amyloid that Mediates Microglial Function. Neuron 97, 1023–1031 e1027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [42].Kang JE, Lim MM, Bateman RJ, Lee JJ, Smyth LP, Cirrito JR, Fujiki N, Nishino S, Holtzman DM (2009) Amyloid-beta dynamics are regulated by orexin and the sleep-wake cycle. Science 326, 1005–1007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [43].Ozcan GG, Lim S, Leighton P, Allison WT, Rihel J (2020) Sleep is bi-directionally modified by amyloid beta oligomers. Elife 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [44].Del Gallo F, Bianchi S, Bertani I, Messa M, Colombo L, Balducci C, Salmona M, Imeri L, Chiesa R (2021) Sleep inhibition induced by amyloid-beta oligomers is mediated by the cellular prion protein. J Sleep Res 30, e13187. [DOI] [PubMed] [Google Scholar]
  • [45].Tobler I, Gaus SE, Deboer T, Achermann P, Fischer M, Rulicke T, Moser M, Oesch B, McBride PA, Manson JC (1996) Altered circadian activity rhythms and sleep in mice devoid of prion protein. Nature 380, 639–642. [DOI] [PubMed] [Google Scholar]
  • [46].Legname G (2017) Elucidating the function of the prion protein. PLoS Pathog 13, e1006458. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [47].Ahnaou A, Raeymaekers L, Steckler T, Drinkenbrug WH (2015) Relevance of the metabotropic glutamate receptor (mGluR5) in the regulation of NREM-REM sleep cycle and homeostasis: evidence from mGluR5 (−/−) mice. Behav Brain Res 282, 218–226. [DOI] [PubMed] [Google Scholar]
  • [48].Holst SC, Sousek A, Hefti K, Saberi-Moghadam S, Buck A, Ametamey SM, Scheidegger M, Franken P, Henning A, Seifritz E, Tafti M, Landolt HP (2017) Cerebral mGluR5 availability contributes to elevated sleep need and behavioral adjustment after sleep deprivation. Elife 6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [49].Brown DR, Nicholas RS, Canevari L (2002) Lack of prion protein expression results in a neuronal phenotype sensitive to stress. J Neurosci Res 67, 211–224. [DOI] [PubMed] [Google Scholar]
  • [50].Pradines E, Loubet D, Mouillet-Richard S, Manivet P, Launay JM, Kellermann O, Schneider B (2009) Cellular prion protein coupling to TACE-dependent TNF-alpha shedding controls neurotransmitter catabolism in neuronal cells. J Neurochem 110, 912–923. [DOI] [PubMed] [Google Scholar]
  • [51].Diering GH, Nirujogi RS, Roth RH, Worley PF, Pandey A, Huganir RL (2017) Homer1a drives homeostatic scaling-down of excitatory synapses during sleep. Science 355, 511–515. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [52].Hu JH, Park JM, Park S, Xiao B, Dehoff MH, Kim S, Hayashi T, Schwarz MK, Huganir RL, Seeburg PH, Linden DJ, Worley PF (2010) Homeostatic scaling requires group I mGluR activation mediated by Homer1a. Neuron 68, 1128–1142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [53].Maret S, Dorsaz S, Gurcel L, Pradervand S, Petit B, Pfister C, Hagenbuchle O, O’Hara BF, Franken P, Tafti M (2007) Homer1a is a core brain molecular correlate of sleep loss. Proc Natl Acad Sci U S A 104, 20090–20095. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [54].Chokshi V, Druciak B, Worley PF, Lee HK (2019) Homer1a Is Required for Establishment of Contralateral Bias and Maintenance of Ocular Dominance in Mouse Visual Cortex. J Neurosci 39, 3897–3905. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [55].Minopoli G, Stante M, Napolitano F, Telese F, Aloia L, De Felice M, Di Lauro R, Pacelli R, Brunetti A, Zambrano N, Russo T (2007) Essential roles for Fe65, Alzheimer amyloid precursor-binding protein, in the cellular response to DNA damage. J Biol Chem 282, 831–835. [DOI] [PubMed] [Google Scholar]
  • [56].Kimberly WT, Zheng JB, Guenette SY, Selkoe DJ (2001) The intracellular domain of the beta-amyloid precursor protein is stabilized by Fe65 and translocates to the nucleus in a notch-like manner. J Biol Chem 276, 40288–40292. [DOI] [PubMed] [Google Scholar]
  • [57].Cao X, Sudhof TC (2001) A transcriptionally [correction of transcriptively] active complex of APP with Fe65 and histone acetyltransferase Tip60. Science 293, 115–120. [DOI] [PubMed] [Google Scholar]
  • [58].Nakaya T, Kawai T, Suzuki T (2008) Regulation of FE65 nuclear translocation and function by amyloid beta-protein precursor in osmotically stressed cells. J Biol Chem 283, 19119–19131. [DOI] [PubMed] [Google Scholar]
  • [59].Koistinen NA, Edlund AK, Menon PK, Ivanova EV, Bacanu S, Iverfeldt K (2017) Nuclear localization of amyloid-beta precursor protein-binding protein Fe65 is dependent on regulated intramembrane proteolysis. PLoS One 12, e0173888. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [60].Kinoshita A, Whelan CM, Smith CJ, Berezovska O, Hyman BT (2002) Direct visualization of the gamma secretase-generated carboxyl-terminal domain of the amyloid precursor protein: association with Fe65 and translocation to the nucleus. J Neurochem 82, 839–847. [DOI] [PubMed] [Google Scholar]
  • [61].Stante M, Minopoli G, Passaro F, Raia M, Vecchio LD, Russo T (2009) Fe65 is required for Tip60-directed histone H4 acetylation at DNA strand breaks. Proc Natl Acad Sci U S A 106, 5093–5098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [62].Murr R, Loizou JI, Yang YG, Cuenin C, Li H, Wang ZQ, Herceg Z (2006) Histone acetylation by Trrap-Tip60 modulates loading of repair proteins and repair of DNA double-strand breaks. Nat Cell Biol 8, 91–99. [DOI] [PubMed] [Google Scholar]
  • [63].von Rotz RC, Kohli BM, Bosset J, Meier M, Suzuki T, Nitsch RM, Konietzko U (2004) The APP intracellular domain forms nuclear multiprotein complexes and regulates the transcription of its own precursor. J Cell Sci 117, 4435–4448. [DOI] [PubMed] [Google Scholar]
  • [64].Schrotter A, Mastalski T, Nensa FM, Neumann M, Loosse C, Pfeiffer K, Magraoui FE, Platta HW, Erdmann R, Theiss C, Uszkoreit J, Eisenacher M, Meyer HE, Marcus K, Muller T (2013) FE65 regulates and interacts with the Bloom syndrome protein in dynamic nuclear spheres - potential relevance to Alzheimer’s disease. J Cell Sci 126, 2480–2492. [DOI] [PubMed] [Google Scholar]
  • [65].Gravel S, Chapman JR, Magill C, Jackson SP (2008) DNA helicases Sgs1 and BLM promote DNA double-strand break resection. Genes Dev 22, 2767–2772. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [66].Cohen S, Guenole A, Lazar I, Marnef A, Clouaire T, Vernekar DV, Puget N, Rocher V, Arnould C, Aguirrebengoa M, Genais M, Firmin N, Shamanna RA, Mourad R, Bohr VA, Borde V, Legube G (2022) A POLD3/BLM dependent pathway handles DSBs in transcribed chromatin upon excessive RNA:DNA hybrid accumulation. Nat Commun 13, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [67].Ward MW, Concannon CG, Whyte J, Walsh CM, Corley B, Prehn JH (2010) The amyloid precursor protein intracellular domain(AICD) disrupts actin dynamics and mitochondrial bioenergetics. J Neurochem 113, 275–284. [DOI] [PubMed] [Google Scholar]
  • [68].Yamazaki Y, Zhao N, Caulfield TR, Liu CC, Bu G (2019) Apolipoprotein E and Alzheimer disease: pathobiology and targeting strategies. Nat Rev Neurol 15, 501–518. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [69].Shi Y, Holtzman DM (2018) Interplay between innate immunity and Alzheimer disease: APOE and TREM2 in the spotlight. Nat Rev Immunol 18, 759–772. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [70].Corder EH, Saunders AM, Strittmatter WJ, Schmechel DE, Gaskell PC, Small GW, Roses AD, Haines JL, Pericak-Vance MA (1993) Gene dose of apolipoprotein E type 4 allele and the risk of Alzheimer’s disease in late onset families. Science 261, 921–923. [DOI] [PubMed] [Google Scholar]
  • [71].Farrer LA, Cupples LA, Haines JL, Hyman B, Kukull WA, Mayeux R, Myers RH, Pericak-Vance MA, Risch N, van Duijn CM (1997) Effects of age, sex, and ethnicity on the association between apolipoprotein E genotype and Alzheimer disease. A meta-analysis. APOE and Alzheimer Disease Meta Analysis Consortium. JAMA 278, 1349–1356. [PubMed] [Google Scholar]
  • [72].Liu L, Aboud O, Jones RA, Mrak RE, Griffin WS, Barger SW (2011) Apolipoprotein E expression is elevated by interleukin 1 and other interleukin 1-induced factors. J Neuroinflammation 8, 175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [73].Mahley RW, Huang Y (2012) Apolipoprotein e sets the stage: response to injury triggers neuropathology. Neuron 76, 871–885. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [74].Xu Q, Bernardo A, Walker D, Kanegawa T, Mahley RW, Huang Y (2006) Profile and regulation of apolipoprotein E (ApoE) expression in the CNS in mice with targeting of green fluorescent protein gene to the ApoE locus. J Neurosci 26, 4985–4994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [75].Hayashi H, Campenot RB, Vance DE, Vance JE (2007) Apolipoprotein E-containing lipoproteins protect neurons from apoptosis via a signaling pathway involving low-density lipoprotein receptor-related protein-1. J Neurosci 27, 1933–1941. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [76].Buttini M, Masliah E, Yu GQ, Palop JJ, Chang S, Bernardo A, Lin C, Wyss-Coray T, Huang Y, Mucke L (2010) Cellular source of apolipoprotein E4 determines neuronal susceptibility to excitotoxic injury in transgenic mice. Am J Pathol 177, 563–569. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [77].Griffin EF, Scopel SE, Stephen CA, Holzhauer AC, Vaji MA, Tuckey RA, Berkowitz LA, Caldwell KA, Caldwell GA (2019) ApoE-associated modulation of neuroprotection from Abeta-mediated neurodegeneration in transgenic Caenorhabditis elegans. Dis Model Mech 12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [78].Ramakrishna S, Jhaveri V, Konings SC, Nawalpuri B, Chakraborty S, Holst B, Schmid B, Gouras GK, Freude KK, Muddashetty RS (2021) APOE4 Affects Basal and NMDAR-Mediated Protein Synthesis in Neurons by Perturbing Calcium Homeostasis. J Neurosci 41, 8686–8709. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [79].Lu K, Nicholas JM, Pertzov Y, Grogan J, Husain M, Pavisic IM, James SN, Parker TD, Lane CA, Keshavan A, Keuss SE, Buchanan SM, Murray-Smith H, Cash DM, Malone IB, Sudre CH, Coath W, Wong A, Henley SMD, Fox NC, Richards M, Schott JM, Crutch SJ (2021) Dissociable effects of APOE-epsilon4 and beta-amyloid pathology on visual working memory. Nat Aging 1, 1002–1009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [80].Zhao N, Liu CC, Van Ingelgom AJ, Martens YA, Linares C, Knight JA, Painter MM, Sullivan PM, Bu G (2017) Apolipoprotein E4 Impairs Neuronal Insulin Signaling by Trapping Insulin Receptor in the Endosomes. Neuron 96, 115–129 e115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [81].Brecht WJ, Harris FM, Chang S, Tesseur I, Yu GQ, Xu Q, Dee Fish J, Wyss-Coray T, Buttini M, Mucke L, Mahley RW, Huang Y (2004) Neuron-specific apolipoprotein e4 proteolysis is associated with increased tau phosphorylation in brains of transgenic mice. J Neurosci 24, 2527–2534. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [82].Narayan P, Orte A, Clarke RW, Bolognesi B, Hook S, Ganzinger KA, Meehan S, Wilson MR, Dobson CM, Klenerman D (2011) The extracellular chaperone clusterin sequesters oligomeric forms of the amyloid-beta(1–40) peptide. Nat Struct Mol Biol 19, 79–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [83].Zhou B, Lu JG, Siddu A, Wernig M, Sudhof TC (2022) Synaptogenic effect of APP-Swedish mutation in familial Alzheimer’s disease. Sci Transl Med 14, eabn9380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [84].Giuffrida ML, Caraci F, Pignataro B, Cataldo S, De Bona P, Bruno V, Molinaro G, Pappalardo G, Messina A, Palmigiano A, Garozzo D, Nicoletti F, Rizzarelli E, Copani A (2009) Beta-amyloid monomers are neuroprotective. J Neurosci 29, 10582–10587. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [85].Plant LD, Boyle JP, Smith IF, Peers C, Pearson HA (2003) The production of amyloid beta peptide is a critical requirement for the viability of central neurons. J Neurosci 23, 5531–5535. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [86].Ramsden M, Henderson Z, Pearson HA (2002) Modulation of Ca2+ channel currents in primary cultures of rat cortical neurones by amyloid beta protein (1-40) is dependent on solubility status. Brain Res 956, 254–261. [DOI] [PubMed] [Google Scholar]
  • [87].Fogel H, Frere S, Segev O, Bharill S, Shapira I, Gazit N, O’Malley T, Slomowitz E, Berdichevsky Y, Walsh DM, Isacoff EY, Hirsch JA, Slutsky I (2014) APP homodimers transduce an amyloid-beta-mediated increase in release probability at excitatory synapses. Cell Rep 7, 1560–1576. [DOI] [PubMed] [Google Scholar]
  • [88].Lazarevic V, Fienko S, Andres-Alonso M, Anni D, Ivanova D, Montenegro-Venegas C, Gundelfinger ED, Cousin MA, Fejtova A (2017) Physiological Concentrations of Amyloid Beta Regulate Recycling of Synaptic Vesicles via Alpha7 Acetylcholine Receptor and CDK5/Calcineurin Signaling. Front Mol Neurosci 10, 221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [89].Gulisano W, Melone M, Ripoli C, Tropea MR, Li Puma DD, Giunta S, Cocco S, Marcotulli D, Origlia N, Palmeri A, Arancio O, Conti F, Grassi C, Puzzo D (2019) Neuromodulatory Action of Picomolar Extracellular Abeta42 Oligomers on Presynaptic and Postsynaptic Mechanisms Underlying Synaptic Function and Memory. J Neurosci 39, 5986–6000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [90].Gulisano W, Melone M, Li Puma DD, Tropea MR, Palmeri A, Arancio O, Grassi C, Conti F, Puzzo D (2018) The effect of amyloid-beta peptide on synaptic plasticity and memory is influenced by different isoforms, concentrations, and aggregation status. Neurobiol Aging 71, 51–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [91].Palmeri A, Ricciarelli R, Gulisano W, Rivera D, Rebosio C, Calcagno E, Tropea MR, Conti S, Das U, Roy S, Pronzato MA, Arancio O, Fedele E, Puzzo D (2017) Amyloid-beta Peptide Is Needed for cGMP-Induced Long-Term Potentiation and Memory. J Neurosci 37, 6926–6937. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [92].Garcia-Osta A, Alberini CM (2009) Amyloid beta mediates memory formation. Learn Mem 16, 267–272. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [93].Morley JE, Farr SA, Banks WA, Johnson SN, Yamada KA, Xu L (2010) A physiological role for amyloid-beta protein:enhancement of learning and memory. J Alzheimers Dis 19, 441–449. [DOI] [PubMed] [Google Scholar]
  • [94].Puzzo D, Privitera L, Leznik E, Fa M, Staniszewski A, Palmeri A, Arancio O (2008) Picomolar amyloid-beta positively modulates synaptic plasticity and memory in hippocampus. J Neurosci 28, 14537–14545. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [95].Barger SW, Harmon AD (1997) Microglial activation by Alzheimer amyloid precursor protein and modulation by apolipoprotein E. Nature 388, 878–881. [DOI] [PubMed] [Google Scholar]
  • [96].Baitsch D, Bock HH, Engel T, Telgmann R, Muller-Tidow C, Varga G, Bot M, Herz J, Robenek H, von Eckardstein A, Nofer JR (2011) Apolipoprotein E induces antiinflammatory phenotype in macrophages. Arterioscler Thromb Vasc Biol 31, 1160–1168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [97].Ali K, Middleton M, Pure E, Rader DJ (2005) Apolipoprotein E suppresses the type I inflammatory response in vivo. Circ Res 97, 922–927. [DOI] [PubMed] [Google Scholar]
  • [98].Waltmann MD, Basford JE, Konaniah ES, Weintraub NL, Hui DY (2014) Apolipoprotein E receptor-2 deficiency enhances macrophage susceptibility to lipid accumulation and cell death to augment atherosclerotic plaque progression and necrosis. Biochim Biophys Acta 1842, 1395–1405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [99].Gordon EM, Yao X, Xu H, Karkowsky W, Kaler M, Kalchiem-Dekel O, Barochia AV, Gao M, Keeran KJ, Jeffries KR, Levine SJ (2019) Apolipoprotein E is a concentration-dependent pulmonary danger signal that activates the NLRP3 inflammasome and IL-1beta secretion by bronchoalveolar fluid macrophages from asthmatic subjects. J Allergy Clin Immunol 144, 426–441 e423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [100].Guo L, LaDu MJ, Van Eldik LJ (2004) A dual role for apolipoprotein e in neuroinflammation: anti- and pro-inflammatory activity. J Mol Neurosci 23, 205–212. [DOI] [PubMed] [Google Scholar]
  • [101].Vainchtein ID, Chin G, Cho FS, Kelley KW, Miller JG, Chien EC, Liddelow SA, Nguyen PT, Nakao-Inoue H, Dorman LC, Akil O, Joshita S, Barres BA, Paz JT, Molofsky AB, Molofsky AV (2018) Astrocyte-derived interleukin-33 promotes microglial synapse engulfment and neural circuit development. Science 359, 1269–1273. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [102].Fu XQ, Peng J, Wang AH, Luo ZG (2020) Tumor necrosis factor alpha mediates neuromuscular synapse elimination. Cell Discov 6, 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [103].Marik SA, Olsen O, Tessier-Lavigne M, Gilbert CD (2013) Death receptor 6 regulates adult experience-dependent cortical plasticity. J Neurosci 33, 14998–15003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [104].Jiang Q, Lee CY, Mandrekar S, Wilkinson B, Cramer P, Zelcer N, Mann K, Lamb B, Willson TM, Collins JL, Richardson JC, Smith JD, Comery TA, Riddell D, Holtzman DM, Tontonoz P, Landreth GE (2008) ApoE promotes the proteolytic degradation of Abeta. Neuron 58, 681–693. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [105].Lee CY, Tse W, Smith JD, Landreth GE (2012) Apolipoprotein E promotes beta-amyloid trafficking and degradation by modulating microglial cholesterol levels. J Biol Chem 287, 2032–2044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [106].Koistinaho M, Lin S, Wu X, Esterman M, Koger D, Hanson J, Higgs R, Liu F, Malkani S, Bales KR, Paul SM (2004) Apolipoprotein E promotes astrocyte colocalization and degradation of deposited amyloid-beta peptides. Nat Med 10, 719–726. [DOI] [PubMed] [Google Scholar]
  • [107].Yeh FL, Wang Y, Tom I, Gonzalez LC, Sheng M (2016) TREM2 Binds to Apolipoproteins, Including APOE and CLU/APOJ, and Thereby Facilitates Uptake of Amyloid-Beta by Microglia. Neuron 91, 328–340. [DOI] [PubMed] [Google Scholar]
  • [108].Atagi Y, Liu CC, Painter MM, Chen XF, Verbeeck C, Zheng H, Li X, Rademakers R, Kang SS, Xu H, Younkin S, Das P, Fryer JD, Bu G (2015) Apolipoprotein E Is a Ligand for Triggering Receptor Expressed on Myeloid Cells 2 (TREM2). J Biol Chem 290, 26043–26050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [109].Bailey CC, DeVaux LB, Farzan M (2015) The Triggering Receptor Expressed on Myeloid Cells 2 Binds Apolipoprotein E. J Biol Chem 290, 26033–26042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [110].Turnbull IR, Gilfillan S, Cella M, Aoshi T, Miller M, Piccio L, Hernandez M, Colonna M (2006) Cutting edge: TREM-2 attenuates macrophage activation. J Immunol 177, 3520–3524. [DOI] [PubMed] [Google Scholar]
  • [111].Andreone BJ, Przybyla L, Llapashtica C, Rana A, Davis SS, van Lengerich B, Lin K, Shi J, Mei Y, Astarita G, Di Paolo G, Sandmann T, Monroe KM, Lewcock JW (2020) Alzheimer’s-associated PLCgamma2 is a signaling node required for both TREM2 function and the inflammatory response in human microglia. Nat Neurosci 23, 927–938. [DOI] [PubMed] [Google Scholar]
  • [112].Nugent AA, Lin K, van Lengerich B, Lianoglou S, Przybyla L, Davis SS, Llapashtica C, Wang J, Kim DJ, Xia D, Lucas A, Baskaran S, Haddick PCG, Lenser M, Earr TK, Shi J, Dugas JC, Andreone BJ, Logan T, Solanoy HO, Chen H, Srivastava A, Poda SB, Sanchez PE, Watts RJ, Sandmann T, Astarita G, Lewcock JW, Monroe KM, Di Paolo G (2020) TREM2 Regulates Microglial Cholesterol Metabolism upon Chronic Phagocytic Challenge. Neuron 105, 837–854 e839. [DOI] [PubMed] [Google Scholar]
  • [113].Koffie RM, Hashimoto T, Tai HC, Kay KR, Serrano-Pozo A, Joyner D, Hou S, Kopeikina KJ, Frosch MP, Lee VM, Holtzman DM, Hyman BT, Spires-Jones TL (2012) Apolipoprotein E4 effects in Alzheimer’s disease are mediated by synaptotoxic oligomeric amyloid-beta. Brain 135, 2155–2168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [114].Ferrari-Souza JP, Lussier FZ, Leffa DT, Therriault J, Tissot C, Bellaver B, Ferreira PCL, Malpetti M, Wang YT, Povala G, Benedet AL, Ashton NJ, Chamoun M, Servaes S, Bezgin G, Kang MS, Stevenson J, Rahmouni N, Pallen V, Poltronetti NM, O’Brien JT, Rowe JB, Cohen AD, Lopez OL, Tudorascu DL, Karikari TK, Klunk WE, Villemagne VL, Soucy JP, Gauthier S, Souza DO, Zetterberg H, Blennow K, Zimmer ER, Rosa-Neto P, Pascoal TA (2023) APOEepsilon4 associates with microglial activation independently of Abeta plaques and tau tangles. Sci Adv 9, eade1474. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [115].Castellano JM, Kim J, Stewart FR, Jiang H, DeMattos RB, Patterson BW, Fagan AM, Morris JC, Mawuenyega KG, Cruchaga C, Goate AM, Bales KR, Paul SM, Bateman RJ, Holtzman DM (2011) Human apoE isoforms differentially regulate brain amyloid-beta peptide clearance. Sci Transl Med 3, 89ra57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [116].Haney MS, Palovics R, Munson CN, Long C, Johansson P, Yip O, Dong W, Rawat E, West E, Schlachetzki JC, Tsai A, Guldner IH, Lamichhane BS, Smith A, Schaum N, Calcuttawala K, Shin A, Wang YH, Wang C, Koutsodendris N, Serrano GE, Beach TG, Reiman EM, Glass CK, Abu-Remaileh M, Enejder A, Huang Y, Wyss-Coray T (2023) APOE4/4 is linked to damaging lipid droplets in Alzheimer’s microglia. bioRxiv. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [117].Lee S, Williams HC, Gorman AA, Devanney NA, Harrison DA, Walsh AE, Goulding DS, Tuck T, Schwartz JL, Zajac DJ, Macauley SL, Estus S, Julia T, Johnson LA, Morganti JM (2023) APOE4 drives transcriptional heterogeneity and maladaptive immunometabolic responses of astrocytes. bioRxiv. [Google Scholar]
  • [118].Victor MB, Leary N, Luna X, Meharena HS, Scannail AN, Bozzelli PL, Samaan G, Murdock MH, von Maydell D, Effenberger AH, Cerit O, Wen HL, Liu L, Welch G, Bonner M, Tsai LH (2022) Lipid accumulation induced by APOE4 impairs microglial surveillance of neuronal-network activity. Cell Stem Cell 29, 1197–1212 e1198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [119].Tcw J, Qian L, Pipalia NH, Chao MJ, Liang SA, Shi Y, Jain BR, Bertelsen SE, Kapoor M, Marcora E, Sikora E, Andrews EJ, Martini AC, Karch CM, Head E, Holtzman DM, Zhang B, Wang M, Maxfield FR, Poon WW, Goate AM (2022) Cholesterol and matrisome pathways dysregulated in astrocytes and microglia. Cell 185, 2213–2233 e2225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [120].Sienski G, Narayan P, Bonner JM, Kory N, Boland S, Arczewska AA, Ralvenius WT, Akay L, Lockshin E, He L, Milo B, Graziosi A, Baru V, Lewis CA, Kellis M, Sabatini DM, Tsai LH, Lindquist S (2021) APOE4 disrupts intracellular lipid homeostasis in human iPSC-derived glia. Sci Transl Med 13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [121].Lin YT, Seo J, Gao F, Feldman HM, Wen HL, Penney J, Cam HP, Gjoneska E, Raja WK, Cheng J, Rueda R, Kritskiy O, Abdurrob F, Peng Z, Milo B, Yu CJ, Elmsaouri S, Dey D, Ko T, Yankner BA, Tsai LH (2018) APOE4 Causes Widespread Molecular and Cellular Alterations Associated with Alzheimer’s Disease Phenotypes in Human iPSC-Derived Brain Cell Types. Neuron 98, 1141–1154 e1147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [122].Zhao J, Fu Y, Yamazaki Y, Ren Y, Davis MD, Liu CC, Lu W, Wang X, Chen K, Cherukuri Y, Jia L, Martens YA, Job L, Shue F, Nguyen TT, Younkin SG, Graff-Radford NR, Wszolek ZK, Brafman DA, Asmann YW, Ertekin-Taner N, Kanekiyo T, Bu G (2020) APOE4 exacerbates synapse loss and neurodegeneration in Alzheimer’s disease patient iPSC-derived cerebral organoids. Nat Commun 11, 5540. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [123].Klann E, Roberson ED, Knapp LT, Sweatt JD (1998) A role for superoxide in protein kinase C activation and induction of long-term potentiation. J Biol Chem 273, 4516–4522. [DOI] [PubMed] [Google Scholar]
  • [124].Thiels E, Urban NN, Gonzalez-Burgos GR, Kanterewicz BI, Barrionuevo G, Chu CT, Oury TD, Klann E (2000) Impairment of long-term potentiation and associative memory in mice that overexpress extracellular superoxide dismutase. J Neurosci 20, 7631–7639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [125].De Pasquale R, Beckhauser TF, Hernandes MS, Giorgetti Britto LR (2014) LTP and LTD in the visual cortex require the activation of NOX2. J Neurosci 34, 12778–12787. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [126].Oswald MC, Brooks PS, Zwart MF, Mukherjee A, West RJ, Giachello CN, Morarach K, Baines RA, Sweeney ST, Landgraf M (2018) Reactive oxygen species regulate activity-dependent neuronal plasticity in Drosophila. Elife 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [127].Francis-Oliveira J, Vilar Higa GS, Mendonca Munhoz Dati L, Carvalho Shieh I, De Pasquale R (2018) Metaplasticity in the Visual Cortex: Crosstalk Between Visual Experience and Reactive Oxygen Species. J Neurosci 38, 5649–5665. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [128].Yi JH, Kim DH, Piers TM, Kim SC, Whitcomb DJ, Regan P, Cho K (2018) Postsynaptic p47phox regulates long-term depression in the hippocampus. Cell Discov 4, 44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [129].Zhang J, Malik A, Choi HB, Ko RW, Dissing-Olesen L, MacVicar BA (2014) Microglial CR3 activation triggers long-term synaptic depression in the hippocampus via NADPH oxidase. Neuron 82, 195–207. [DOI] [PubMed] [Google Scholar]
  • [130].Reyes RC, Brennan AM, Shen Y, Baldwin Y, Swanson RA (2012) Activation of neuronal NMDA receptors induces superoxide-mediated oxidative stress in neighboring neurons and astrocytes. J Neurosci 32, 12973–12978. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [131].Pollina EA, Gilliam DT, Landau AT, Lin C, Pajarillo N, Davis CP, Harmin DA, Yap EL, Vogel IR, Griffith EC, Nagy MA, Ling E, Duffy EE, Sabatini BL, Weitz CJ, Greenberg ME (2023) A NPAS4-NuA4 complex couples synaptic activity to DNA repair. Nature 614, 732–741. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [132].Delint-Ramirez I, Konada L, Heady L, Rueda R, Jacome ASV, Marlin E, Marchioni C, Segev A, Kritskiy O, Yamakawa S, Reiter AH, Tsai LH, Madabhushi R (2022) Calcineurin dephosphorylates topoisomerase IIbeta and regulates the formation of neuronal-activity-induced DNA breaks. Mol Cell 82, 3794–3809 e3798. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [133].Madabhushi R, Gao F, Pfenning AR, Pan L, Yamakawa S, Seo J, Rueda R, Phan TX, Yamakawa H, Pao PC, Stott RT, Gjoneska E, Nott A, Cho S, Kellis M, Tsai LH (2015) Activity-Induced DNA Breaks Govern the Expression of Neuronal Early-Response Genes. Cell 161, 1592–1605. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [134].Suberbielle E, Sanchez PE, Kravitz AV, Wang X, Ho K, Eilertson K, Devidze N, Kreitzer AC, Mucke L (2013) Physiologic brain activity causes DNA double-strand breaks in neurons, with exacerbation by amyloid-beta. Nat Neurosci 16, 613–621. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [135].Almenar-Queralt A, Falzone TL, Shen Z, Lillo C, Killian RL, Arreola AS, Niederst ED, Ng KS, Kim SN, Briggs SP, Williams DS, Goldstein LS (2014) UV irradiation accelerates amyloid precursor protein (APP) processing and disrupts APP axonal transport. J Neurosci 34, 3320–3339. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [136].Ma S, Rong Z, Liu C, Qin X, Zhang X, Chen Q (2021) DNA damage promotes microtubule dynamics through a DNA-PK-AKT axis for enhanced repair. J Cell Biol 220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [137].Lottersberger F, Karssemeijer RA, Dimitrova N, de Lange T (2015) 53BP1 and the LINC Complex Promote Microtubule-Dependent DSB Mobility and DNA Repair. Cell 163, 880–893. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [138].Oshidari R, Strecker J, Chung DKC, Abraham KJ, Chan JNY, Damaren CJ, Mekhail K (2018) Nuclear microtubule filaments mediate non-linear directional motion of chromatin and promote DNA repair. Nat Commun 9, 2567. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [139].Sultan A, Nesslany F, Violet M, Begard S, Loyens A, Talahari S, Mansuroglu Z, Marzin D, Sergeant N, Humez S, Colin M, Bonnefoy E, Buee L, Galas MC (2011) Nuclear tau, a key player in neuronal DNA protection. J Biol Chem 286, 4566–4575. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [140].Asada-Utsugi M, Uemura K, Ayaki T, M TU, Minamiyama S, Hikiami R, Morimura T, Shodai A, Ueki T, Takahashi R, Kinoshita A, Urushitani M (2022) Failure of DNA double-strand break repair by tau mediates Alzheimer’s disease pathology in vitro. Commun Biol 5, 358. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [141].Lester E, Ooi FK, Bakkar N, Ayers J, Woerman AL, Wheeler J, Bowser R, Carlson GA, Prusiner SB, Parker R (2021) Tau aggregates are RNA-protein assemblies that mislocalize multiple nuclear speckle components. Neuron 109, 1675–1691 e1679. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [142].Montalbano M, McAllen S, Puangmalai N, Sengupta U, Bhatt N, Johnson OD, Kharas MG, Kayed R (2020) RNA-binding proteins Musashi and tau soluble aggregates initiate nuclear dysfunction. Nat Commun 11, 4305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [143].Roqanian S, Ahmadian S, Nabavi SM, Pakdaman H, Shafiezadeh M, Goudarzi G, Shahpasand K (2022) Tau nuclear translocation is a leading step in tau pathology process through P53 stabilization and nucleolar dispersion. J Neurosci Res 100, 1084–1104. [DOI] [PubMed] [Google Scholar]
  • [144].Bengoa-Vergniory N, Velentza-Almpani E, Silva AM, Scott C, Vargas-Caballero M, Sastre M, Wade-Martins R, Alegre-Abarrategui J (2021) Tau-proximity ligation assay reveals extensive previously undetected pathology prior to neurofibrillary tangles in preclinical Alzheimer’s disease. Acta Neuropathol Commun 9, 18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [145].Gil L, Nino SA, Chi-Ahumada E, Rodriguez-Leyva I, Guerrero C, Rebolledo AB, Arias JA, Jimenez-Capdeville ME (2020) Perinuclear Lamin A and Nucleoplasmic Lamin B2 Characterize Two Types of Hippocampal Neurons through Alzheimer’s Disease Progression. Int J Mol Sci 21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [146].Watcharasit P, Bijur GN, Zmijewski JW, Song L, Zmijewska A, Chen X, Johnson GV, Jope RS (2002) Direct, activating interaction between glycogen synthase kinase-3beta and p53 after DNA damage. Proc Natl Acad Sci U S A 99, 7951–7955. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [147].Iijima-Ando K, Zhao L, Gatt A, Shenton C, Iijima K (2010) A DNA damage-activated checkpoint kinase phosphorylates tau and enhances tau-induced neurodegeneration. Hum Mol Genet 19, 1930–1938. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [148].Bulavin DV, Higashimoto Y, Popoff IJ, Gaarde WA, Basrur V, Potapova O, Appella E, Fornace AJ Jr. (2001) Initiation of a G2/M checkpoint after ultraviolet radiation requires p38 kinase. Nature 411, 102–107. [DOI] [PubMed] [Google Scholar]
  • [149].Morales AJ, Carrero JA, Hung PJ, Tubbs AT, Andrews JM, Edelson BT, Calderon B, Innes CL, Paules RS, Payton JE, Sleckman BP (2017) A type I IFN-dependent DNA damage response regulates the genetic program and inflammasome activation in macrophages. Elife 6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [150].Bednarski JJ, Sleckman BP (2019) At the intersection of DNA damage and immune responses. Nat Rev Immunol 19, 231–242. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [151].Yang D, Elner SG, Bian ZM, Till GO, Petty HR, Elner VM (2007) Pro-inflammatory cytokines increase reactive oxygen species through mitochondria and NADPH oxidase in cultured RPE cells. Exp Eye Res 85, 462–472. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [152].Jin S, Ray RM, Johnson LR (2008) TNF-alpha/cycloheximide-induced apoptosis in intestinal epithelial cells requires Rac1-regulated reactive oxygen species. Am J Physiol Gastrointest Liver Physiol 294, G928–937. [DOI] [PubMed] [Google Scholar]
  • [153].Druzhyna NM, Musiyenko SI, Wilson GL, LeDoux SP (2005) Cytokines induce nitric oxide-mediated mtDNA damage and apoptosis in oligodendrocytes. Protective role of targeting 8-oxoguanine glycosylase to mitochondria. J Biol Chem 280, 21673–21679. [DOI] [PubMed] [Google Scholar]
  • [154].Yakes FM, Van Houten B (1997) Mitochondrial DNA damage is more extensive and persists longer than nuclear DNA damage in human cells following oxidative stress. Proc Natl Acad Sci U S A 94, 514–519. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [155].Samavati L, Lee I, Mathes I, Lottspeich F, Huttemann M (2008) Tumor necrosis factor alpha inhibits oxidative phosphorylation through tyrosine phosphorylation at subunit I of cytochrome c oxidase. J Biol Chem 283, 21134–21144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [156].Kim J, Xu M, Xo R, Mates A, Wilson GL, Pearsall AWt, Grishko V (2010) Mitochondrial DNA damage is involved in apoptosis caused by pro-inflammatory cytokines in human OA chondrocytes. Osteoarthritis Cartilage 18, 424–432. [DOI] [PubMed] [Google Scholar]
  • [157].Lustbader JW, Cirilli M, Lin C, Xu HW, Takuma K, Wang N, Caspersen C, Chen X, Pollak S, Chaney M, Trinchese F, Liu S, Gunn-Moore F, Lue LF, Walker DG, Kuppusamy P, Zewier ZL, Arancio O, Stern D, Yan SS, Wu H (2004) ABAD directly links Abeta to mitochondrial toxicity in Alzheimer’s disease. Science 304, 448–452. [DOI] [PubMed] [Google Scholar]
  • [158].Yao J, Du H, Yan S, Fang F, Wang C, Lue LF, Guo L, Chen D, Stern DM, Gunn Moore FJ, Xi Chen J, Arancio O, Yan SS (2011) Inhibition of amyloid-beta (Abeta) peptide-binding alcohol dehydrogenase-Abeta interaction reduces Abeta accumulation and improves mitochondrial function in a mouse model of Alzheimer’s disease. J Neurosci 31, 2313–2320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [159].Hernandez-Zimbron LF, Luna-Munoz J, Mena R, Vazquez-Ramirez R, Kubli-Garfias C, Cribbs DH, Manoutcharian K, Gevorkian G (2012) Amyloid-beta peptide binds to cytochrome C oxidase subunit 1. PLoS One 7, e42344. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [160].Beck SJ, Guo L, Phensy A, Tian J, Wang L, Tandon N, Gauba E, Lu L, Pascual JM, Kroener S, Du H (2016) Deregulation of mitochondrial F1FO-ATP synthase via OSCP in Alzheimer’s disease. Nat Commun 7, 11483. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [161].Lee A, Kondapalli C, Virga DM, Lewis TL Jr, Koo SY, Ashok A, Mairet-Coello G, Herzig S, Foretz M, Viollet B, Shaw R, Sproul A, Polleux F (2022) Abeta42 oligomers trigger synaptic loss through CAMKK2-AMPK-dependent effectors coordinating mitochondrial fission and mitophagy. Nat Commun 13, 4444. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [162].Kim J, Yang Y, Song SS, Na JH, Oh KJ, Jeong C, Yu YG, Shin YK (2014) Beta-amyloid oligomers activate apoptotic BAK pore for cytochrome c release. Biophys J 107, 1601–1608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [163].Sage JM, Gildemeister OS, Knight KL (2010) Discovery of a novel function for human Rad51: maintenance of the mitochondrial genome. J Biol Chem 285, 18984–18990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [164].Mishra A, Saxena S, Kaushal A, Nagaraju G (2018) RAD51C/XRCC3 Facilitates Mitochondrial DNA Replication and Maintains Integrity of the Mitochondrial Genome. Mol Cell Biol 38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [165].Luzwick JW, Dombi E, Boisvert RA, Roy S, Park S, Kunnimalaiyaan S, Goffart S, Schindler D, Schlacher K (2021) MRE11-dependent instability in mitochondrial DNA fork protection activates a cGAS immune signaling pathway. Sci Adv 7, eabf9441. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [166].Klucnika A, Mu P, Jezek J, McCormack M, Di Y, Bradshaw CR, Ma H (2023) REC drives recombination to repair double-strand breaks in animal mtDNA. J Cell Biol 222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [167].Wilkins HM (2023) Interactions between amyloid, amyloid precursor protein, and mitochondria. Biochem Soc Trans 51, 173–182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [168].Gulen MF, Samson N, Keller A, Schwabenland M, Liu C, Gluck S, Thacker VV, Favre L, Mangeat B, Kroese LJ, Krimpenfort P, Prinz M, Ablasser A (2023) cGAS-STING drives ageing-related inflammation and neurodegeneration. Nature 620, 374–380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [169].Nidadavolu LS, Feger D, Chen D, Wu Y, Grodstein F, Gross AL, Bennett DA, Walston JD, Oh ES, Abadir PM (2023) Associations between circulating cell-free mitochondrial DNA, inflammatory markers, and cognitive and physical outcomes in community dwelling older adults. Immun Ageing 20, 24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [170].Kim J, Gupta R, Blanco LP, Yang S, Shteinfer-Kuzmine A, Wang K, Zhu J, Yoon HE, Wang X, Kerkhofs M, Kang H, Brown AL, Park SJ, Xu X, Zandee van Rilland E, Kim MK, Cohen JI, Kaplan MJ, Shoshan-Barmatz V, Chung JH (2019) VDAC oligomers form mitochondrial pores to release mtDNA fragments and promote lupus-like disease. Science 366, 1531–1536. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [171].Pinti M, Cevenini E, Nasi M, De Biasi S, Salvioli S, Monti D, Benatti S, Gibellini L, Cotichini R, Stazi MA, Trenti T, Franceschi C, Cossarizza A (2014) Circulating mitochondrial DNA increases with age and is a familiar trait: Implications for “inflamm-aging”. Eur J Immunol 44, 1552–1562. [DOI] [PubMed] [Google Scholar]
  • [172].Hou Y, Wei Y, Lautrup S, Yang B, Wang Y, Cordonnier S, Mattson MP, Croteau DL, Bohr VA (2021) NAD(+) supplementation reduces neuroinflammation and cell senescence in a transgenic mouse model of Alzheimer’s disease via cGAS-STING. Proc Natl Acad Sci U S A 118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [173].Dileep V, Boix CA, Mathys H, Marco A, Welch GM, Meharena HS, Loon A, Jeloka R, Peng Z, Bennett DA, Kellis M, Tsai LH (2023) Neuronal DNA double-strand breaks lead to genome structural variations and 3D genome disruption in neurodegeneration. Cell 186, 4404–4421 e4420. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [174].Mathys H, Peng Z, Boix CA, Victor MB, Leary N, Babu S, Abdelhady G, Jiang X, Ng AP, Ghafari K, Kunisky AK, Mantero J, Galani K, Lohia VN, Fortier GE, Lotfi Y, Ivey J, Brown HP, Patel PR, Chakraborty N, Beaudway JI, Imhoff EJ, Keeler CF, McChesney MM, Patel HH, Patel SP, Thai MT, Bennett DA, Kellis M, Tsai LH (2023) Single-cell atlas reveals correlates of high cognitive function, dementia, and resilience to Alzheimer’s disease pathology. Cell 186, 4365–4385 e4327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [175].Kim T, Vidal GS, Djurisic M, William CM, Birnbaum ME, Garcia KC, Hyman BT, Shatz CJ (2013) Human LilrB2 is a beta-amyloid receptor and its murine homolog PirB regulates synaptic plasticity in an Alzheimer’s model. Science 341, 1399–1404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [176].Syken J, Grandpre T, Kanold PO, Shatz CJ (2006) PirB restricts ocular-dominance plasticity in visual cortex. Science 313, 1795–1800. [DOI] [PubMed] [Google Scholar]
  • [177].Datwani A, McConnell MJ, Kanold PO, Micheva KD, Busse B, Shamloo M, Smith SJ, Shatz CJ (2009) Classical MHCI molecules regulate retinogeniculate refinement and limit ocular dominance plasticity. Neuron 64, 463–470. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [178].Huh GS, Boulanger LM, Du H, Riquelme PA, Brotz TM, Shatz CJ (2000) Functional requirement for class I MHC in CNS development and plasticity. Science 290, 2155–2159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [179].Lauren J, Gimbel DA, Nygaard HB, Gilbert JW, Strittmatter SM (2009) Cellular prion protein mediates impairment of synaptic plasticity by amyloid-beta oligomers. Nature 457, 1128–1132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [180].Um JW, Nygaard HB, Heiss JK, Kostylev MA, Stagi M, Vortmeyer A, Wisniewski T, Gunther EC, Strittmatter SM (2012) Alzheimer amyloid-beta oligomer bound to postsynaptic prion protein activates Fyn to impair neurons. Nat Neurosci 15, 1227–1235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [181].Um JW, Kaufman AC, Kostylev M, Heiss JK, Stagi M, Takahashi H, Kerrisk ME, Vortmeyer A, Wisniewski T, Koleske AJ, Gunther EC, Nygaard HB, Strittmatter SM (2013) Metabotropic glutamate receptor 5 is a coreceptor for Alzheimer abeta oligomer bound to cellular prion protein. Neuron 79, 887–902. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [182].Kostylev MA, Tuttle MD, Lee S, Klein LE, Takahashi H, Cox TO, Gunther EC, Zilm KW, Strittmatter SM (2018) Liquid and Hydrogel Phases of PrP(C) Linked to Conformation Shifts and Triggered by Alzheimer’s Amyloid-beta Oligomers. Mol Cell 72, 426–443 e412. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [183].Heavner WE, Lautz JD, Speed HE, Gniffke EP, Immendorf KB, Welsh JP, Baertsch NA, Smith SEP (2021) Remodeling of the Homer-Shank interactome mediates homeostatic plasticity. Sci Signal 14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [184].Hsieh H, Boehm J, Sato C, Iwatsubo T, Tomita T, Sisodia S, Malinow R (2006) AMPAR removal underlies Abeta-induced synaptic depression and dendritic spine loss. Neuron 52, 831–843. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [185].Hu NW, Nicoll AJ, Zhang D, Mably AJ, O’Malley T, Purro SA, Terry C, Collinge J, Walsh DM, Rowan MJ (2014) mGlu5 receptors and cellular prion protein mediate amyloid-beta-facilitated synaptic long-term depression in vivo. Nat Commun 5, 3374. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [186].Cisse M, Halabisky B, Harris J, Devidze N, Dubal DB, Sun B, Orr A, Lotz G, Kim DH, Hamto P, Ho K, Yu GQ, Mucke L (2011) Reversing EphB2 depletion rescues cognitive functions in Alzheimer model. Nature 469, 47–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [187].Magdesian MH, Carvalho MM, Mendes FA, Saraiva LM, Juliano MA, Juliano L, Garcia-Abreu J, Ferreira ST (2008) Amyloid-beta binds to the extracellular cysteine-rich domain of Frizzled and inhibits Wnt/beta-catenin signaling. J Biol Chem 283, 9359–9368. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [188].Feng B, Freitas AE, Gorodetski L, Wang J, Tian R, Lee YR, Grewal AS, Zou Y (2021) Planar cell polarity signaling components are a direct target of beta-amyloid-associated degeneration of glutamatergic synapses. Sci Adv 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [189].Thomazeau A, Bosch M, Essayan-Perez S, Barnes SA, De Jesus-Cortes H, Bear MF (2021) Dissociation of functional and structural plasticity of dendritic spines during NMDAR and mGluR-dependent long-term synaptic depression in wild-type and fragile X model mice. Mol Psychiatry 26, 4652–4669. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [190].Stein IS, Gray JA, Zito K (2015) Non-Ionotropic NMDA Receptor Signaling Drives Activity-Induced Dendritic Spine Shrinkage. J Neurosci 35, 12303–12308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [191].Kessels HW, Nabavi S, Malinow R (2013) Metabotropic NMDA receptor function is required for beta-amyloid-induced synaptic depression. Proc Natl Acad Sci U S A 110, 4033–4038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [192].Nabavi S, Kessels HW, Alfonso S, Aow J, Fox R, Malinow R (2013) Metabotropic NMDA receptor function is required for NMDA receptor-dependent long-term depression. Proc Natl Acad Sci U S A 110, 4027–4032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [193].Roselli F, Tirard M, Lu J, Hutzler P, Lamberti P, Livrea P, Morabito M, Almeida OF (2005) Soluble beta-amyloid1-40 induces NMDA-dependent degradation of postsynaptic density-95 at glutamatergic synapses. J Neurosci 25, 11061–11070. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [194].Snyder EM, Nong Y, Almeida CG, Paul S, Moran T, Choi EY, Nairn AC, Salter MW, Lombroso PJ, Gouras GK, Greengard P (2005) Regulation of NMDA receptor trafficking by amyloid-beta. Nat Neurosci 8, 1051–1058. [DOI] [PubMed] [Google Scholar]
  • [195].Shankar GM, Bloodgood BL, Townsend M, Walsh DM, Selkoe DJ, Sabatini BL (2007) Natural oligomers of the Alzheimer amyloid-beta protein induce reversible synapse loss by modulating an NMDA-type glutamate receptor-dependent signaling pathway. J Neurosci 27, 2866–2875. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [196].Lacor PN, Buniel MC, Furlow PW, Clemente AS, Velasco PT, Wood M, Viola KL, Klein WL (2007) Abeta oligomer-induced aberrations in synapse composition, shape, and density provide a molecular basis for loss of connectivity in Alzheimer’s disease. J Neurosci 27, 796–807. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [197].Babiec WE, Guglietta R, Jami SA, Morishita W, Malenka RC, O’Dell TJ (2014) Ionotropic NMDA receptor signaling is required for the induction of long-term depression in the mouse hippocampal CA1 region. J Neurosci 34, 5285–5290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [198].Birnbaum JH, Bali J, Rajendran L, Nitsch RM, Tackenberg C (2015) Calcium flux-independent NMDA receptor activity is required for Abeta oligomer-induced synaptic loss. Cell Death Dis 6, e1791. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [199].Zalocusky KA, Najm R, Taubes AL, Hao Y, Yoon SY, Koutsodendris N, Nelson MR, Rao A, Bennett DA, Bant J, Amornkul DJ, Xu Q, An A, Cisne-Thomson O, Huang Y (2021) Neuronal ApoE upregulates MHC-I expression to drive selective neurodegeneration in Alzheimer’s disease. Nat Neurosci 24, 786–798. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [200].Corriveau RA, Huh GS, Shatz CJ (1998) Regulation of class I MHC gene expression in the developing and mature CNS by neural activity. Neuron 21, 505–520. [DOI] [PubMed] [Google Scholar]
  • [201].Lee H, Brott BK, Kirkby LA, Adelson JD, Cheng S, Feller MB, Datwani A, Shatz CJ (2014) Synapse elimination and learning rules co-regulated by MHC class I H2-Db. Nature 509, 195–200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [202].Leonhardt RM, Keusekotten K, Bekpen C, Knittler MR (2005) Critical role for the tapasin-docking site of TAP2 in the functional integrity of the MHC class I-peptide-loading complex. J Immunol 175, 5104–5114. [DOI] [PubMed] [Google Scholar]
  • [203].Hanuscheck N, Thalman C, Domingues M, Schmaul S, Muthuraman M, Hetsch F, Ecker M, Endle H, Oshaghi M, Martino G, Kuhlmann T, Bozek K, van Beers T, Bittner S, von Engelhardt J, Vogt J, Vogelaar CF, Zipp F (2022) Interleukin-4 receptor signaling modulates neuronal network activity. J Exp Med 219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [204].Zhao X, Wang H, Sun G, Zhang J, Edwards NJ, Aronowski J (2015) Neuronal Interleukin-4 as a Modulator of Microglial Pathways and Ischemic Brain Damage. J Neurosci 35, 11281–11291. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [205].Yasuda M, Nagappan-Chettiar S, Johnson-Venkatesh EM, Umemori H (2021) An activity-dependent determinant of synapse elimination in the mammalian brain. Neuron 109, 1333–1349 e1336. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [206].Trusca VG, Fuior EV, Florea IC, Kardassis D, Simionescu M, Gafencu AV (2011) Macrophage-specific up-regulation of apolipoprotein E gene expression by STAT1 is achieved via long range genomic interactions. J Biol Chem 286, 13891–13904. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [207].Leng K, Li E, Eser R, Piergies A, Sit R, Tan M, Neff N, Li SH, Rodriguez RD, Suemoto CK, Leite REP, Ehrenberg AJ, Pasqualucci CA, Seeley WW, Spina S, Heinsen H, Grinberg LT, Kampmann M (2021) Molecular characterization of selectively vulnerable neurons in Alzheimer’s disease. Nat Neurosci 24, 276–287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [208].Jabaudon D, Shnider SJ, Tischfield DJ, Galazo MJ, Macklis JD (2012) RORbeta induces barrel-like neuronal clusters in the developing neocortex. Cereb Cortex 22, 996–1006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [209].Clark EA, Rutlin M, Capano L, Aviles S, Saadon JR, Taneja P, Zhang Q, Bullis JB, Lauer T, Myers E, Schulmann A, Forrest D, Nelson SB (2020) Cortical RORbeta is required for layer 4 transcriptional identity and barrel integrity. Elife 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [210].Wang C, Xiong M, Gratuze M, Bao X, Shi Y, Andhey PS, Manis M, Schroeder C, Yin Z, Madore C, Butovsky O, Artyomov M, Ulrich JD, Holtzman DM (2021) Selective removal of astrocytic APOE4 strongly protects against tau-mediated neurodegeneration and decreases synaptic phagocytosis by microglia. Neuron 109, 1657–1674 e1657. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [211].Li W, Hofer MJ, Songkhunawej P, Jung SR, Hancock D, Denyer G, Campbell IL (2017) Type I interferon-regulated gene expression and signaling in murine mixed glial cells lacking signal transducers and activators of transcription 1 or 2 or interferon regulatory factor 9. J Biol Chem 292, 5845–5859. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [212].Barker SJ, Raju RM, Milman NEP, Wang J, Davila-Velderrain J, Gunter-Rahman F, Parro CC, Bozzelli PL, Abdurrob F, Abdelaal K, Bennett DA, Kellis M, Tsai LH (2021) MEF2 is a key regulator of cognitive potential and confers resilience to neurodegeneration. Sci Transl Med 13, eabd7695. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [213].Udeochu JC, Amin S, Huang Y, Fan L, Torres ERS, Carling GK, Liu B, McGurran H, Coronas-Samano G, Kauwe G, Mousa GA, Wong MY, Ye P, Nagiri RK, Lo I, Holtzman J, Corona C, Yarahmady A, Gill MT, Raju RM, Mok SA, Gong S, Luo W, Zhao M, Tracy TE, Ratan RR, Tsai LH, Sinha SC, Gan L (2023) Tau activation of microglial cGAS-IFN reduces MEF2C-mediated cognitive resilience. Nat Neurosci 26, 737–750. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [214].Xiao F, Wang H, Fu X, Li Y, Ma K, Sun L, Gao X, Wu Z (2011) Oncostatin M inhibits myoblast differentiation and regulates muscle regeneration. Cell Res 21, 350–364. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [215].Luscher C, Huber KM (2010) Group 1 mGluR-dependent synaptic long-term depression: mechanisms and implications for circuitry and disease. Neuron 65, 445–459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [216].Collingridge GL, Peineau S, Howland JG, Wang YT (2010) Long-term depression in the CNS. Nat Rev Neurosci 11, 459–473. [DOI] [PubMed] [Google Scholar]
  • [217].Spacek J, Harris KM (1997) Three-dimensional organization of smooth endoplasmic reticulum in hippocampal CA1 dendrites and dendritic spines of the immature and mature rat. J Neurosci 17, 190–203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [218].Yap K, Drakew A, Smilovic D, Rietsche M, Paul MH, Vuksic M, Del Turco D, Deller T (2020) The actin-modulating protein synaptopodin mediates long-term survival of dendritic spines. Elife 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [219].Okubo-Suzuki R, Okada D, Sekiguchi M, Inokuchi K (2008) Synaptopodin maintains the neural activity-dependent enlargement of dendritic spines in hippocampal neurons. Mol Cell Neurosci 38, 266–276. [DOI] [PubMed] [Google Scholar]
  • [220].Vlachos A, Korkotian E, Schonfeld E, Copanaki E, Deller T, Segal M (2009) Synaptopodin regulates plasticity of dendritic spines in hippocampal neurons. J Neurosci 29, 1017–1033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [221].Deller T, Korte M, Chabanis S, Drakew A, Schwegler H, Stefani GG, Zuniga A, Schwarz K, Bonhoeffer T, Zeller R, Frotscher M, Mundel P (2003) Synaptopodin-deficient mice lack a spine apparatus and show deficits in synaptic plasticity. Proc Natl Acad Sci U S A 100, 10494–10499. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [222].Holbro N, Grunditz A, Oertner TG (2009) Differential distribution of endoplasmic reticulum controls metabotropic signaling and plasticity at hippocampal synapses. Proc Natl Acad Sci U S A 106, 15055–15060. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [223].Speranza L, Inglebert Y, De Sanctis C, Wu PY, Kalinowska M, McKinney RA, Francesconi A (2022) Stabilization of Spine Synaptopodin by mGluR1 Is Required for mGluR-LTD. J Neurosci 42, 1666–1678. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [224].Oh WC, Hill TC, Zito K (2013) Synapse-specific and size-dependent mechanisms of spine structural plasticity accompanying synaptic weakening. Proc Natl Acad Sci U S A 110, E305–312. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [225].Dalva MB, Takasu MA, Lin MZ, Shamah SM, Hu L, Gale NW, Greenberg ME (2000) EphB receptors interact with NMDA receptors and regulate excitatory synapse formation. Cell 103, 945–956. [DOI] [PubMed] [Google Scholar]
  • [226].Hanamura K, Washburn HR, Sheffler-Collins SI, Xia NL, Henderson N, Tillu DV, Hassler S, Spellman DS, Zhang G, Neubert TA, Price TJ, Dalva MB (2017) Extracellular phosphorylation of a receptor tyrosine kinase controls synaptic localization of NMDA receptors and regulates pathological pain. PLoS Biol 15, e2002457. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [227].Grunwald IC, Korte M, Wolfer D, Wilkinson GA, Unsicker K, Lipp HP, Bonhoeffer T, Klein R (2001) Kinase-independent requirement of EphB2 receptors in hippocampal synaptic plasticity. Neuron 32, 1027–1040. [DOI] [PubMed] [Google Scholar]
  • [228].Henderson JT, Georgiou J, Jia Z, Robertson J, Elowe S, Roder JC, Pawson T (2001) The receptor tyrosine kinase EphB2 regulates NMDA-dependent synaptic function. Neuron 32, 1041–1056. [DOI] [PubMed] [Google Scholar]
  • [229].Thakar S, Wang L, Yu T, Ye M, Onishi K, Scott J, Qi J, Fernandes C, Han X, Yates JR 3rd, Berg DK, Zou Y (2017) Evidence for opposing roles of Celsr3 and Vangl2 in glutamatergic synapse formation. Proc Natl Acad Sci U S A 114, E610–E618. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [230].Ban Y, Yu T, Feng B, Lorenz C, Wang X, Baker C, Zou Y (2021) Prickle promotes the formation and maintenance of glutamatergic synapses by stabilizing the intercellular planar cell polarity complex. Sci Adv 7, eabh2974. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [231].Chen B, Wang L, Li X, Shi Z, Duan J, Wei JA, Li C, Pang C, Wang D, Zhang K, Chen H, Na W, Zhang L, So KF, Zhou L, Jiang B, Yuan TF, Qu Y (2022) Celsr2 regulates NMDA receptors and dendritic homeostasis in dorsal CA1 to enable social memory. Mol Psychiatry. [DOI] [PubMed] [Google Scholar]
  • [232].Nagaoka T, Ohashi R, Inutsuka A, Sakai S, Fujisawa N, Yokoyama M, Huang YH, Igarashi M, Kishi M (2014) The Wnt/planar cell polarity pathway component Vangl2 induces synapse formation through direct control of N-cadherin. Cell Rep 6, 916–927. [DOI] [PubMed] [Google Scholar]
  • [233].Corbett GT, Wang Z, Hong W, Colom-Cadena M, Rose J, Liao M, Asfaw A, Hall TC, Ding L, DeSousa A, Frosch MP, Collinge J, Harris DA, Perkinton MS, Spires-Jones TL, Young-Pearse TL, Billinton A, Walsh DM (2020) PrP is a central player in toxicity mediated by soluble aggregates of neurodegeneration-causing proteins. Acta Neuropathol 139, 503–526. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [234].Zou WQ, Xiao X, Yuan J, Puoti G, Fujioka H, Wang X, Richardson S, Zhou X, Zou R, Li S, Zhu X, McGeer PL, McGeehan J, Kneale G, Rincon-Limas DE, Fernandez-Funez P, Lee HG, Smith MA, Petersen RB, Guo JP (2011) Amyloid-beta42 interacts mainly with insoluble prion protein in the Alzheimer brain. J Biol Chem 286, 15095–15105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [235].Dohler F, Sepulveda-Falla D, Krasemann S, Altmeppen H, Schluter H, Hildebrand D, Zerr I, Matschke J, Glatzel M (2014) High molecular mass assemblies of amyloid-beta oligomers bind prion protein in patients with Alzheimer’s disease. Brain 137, 873–886. [DOI] [PubMed] [Google Scholar]
  • [236].Yeung JHY, Walby JL, Palpagama TH, Turner C, Waldvogel HJ, Faull RLM, Kwakowsky A (2021) Glutamatergic receptor expression changes in the Alzheimer’s disease hippocampus and entorhinal cortex. Brain Pathol 31, e13005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [237].Kravitz E, Gaisler-Salomon I, Biegon A (2013) Hippocampal glutamate NMDA receptor loss tracks progression in Alzheimer’s disease: quantitative autoradiography in postmortem human brain. PLoS One 8, e81244. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [238].Sze C, Bi H, Kleinschmidt-DeMasters BK, Filley CM, Martin LJ (2001) N-Methyl-D-aspartate receptor subunit proteins and their phosphorylation status are altered selectively in Alzheimer’s disease. J Neurol Sci 182, 151–159. [DOI] [PubMed] [Google Scholar]
  • [239].Ikonomovic MD, Mizukami K, Warde D, Sheffield R, Hamilton R, Wenthold RJ, Armstrong DM (1999) Distribution of glutamate receptor subunit NMDAR1 in the hippocampus of normal elderly and patients with Alzheimer’s disease. Exp Neurol 160, 194–204. [DOI] [PubMed] [Google Scholar]
  • [240].Koopman G, Reutelingsperger CP, Kuijten GA, Keehnen RM, Pals ST, van Oers MH (1994) Annexin V for flow cytometric detection of phosphatidylserine expression on B cells undergoing apoptosis. Blood 84, 1415–1420. [PubMed] [Google Scholar]
  • [241].Martin SJ, Reutelingsperger CP, McGahon AJ, Rader JA, van Schie RC, LaFace DM, Green DR (1995) Early redistribution of plasma membrane phosphatidylserine is a general feature of apoptosis regardless of the initiating stimulus: inhibition by overexpression of Bcl-2 and Abl. J Exp Med 182, 1545–1556. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [242].Scott-Hewitt N, Perrucci F, Morini R, Erreni M, Mahoney M, Witkowska A, Carey A, Faggiani E, Schuetz LT, Mason S, Tamborini M, Bizzotto M, Passoni L, Filipello F, Jahn R, Stevens B, Matteoli M (2020) Local externalization of phosphatidylserine mediates developmental synaptic pruning by microglia. EMBO J 39, e105380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [243].Li T, Chiou B, Gilman CK, Luo R, Koshi T, Yu D, Oak HC, Giera S, Johnson-Venkatesh E, Muthukumar AK, Stevens B, Umemori H, Piao X (2020) A splicing isoform of GPR56 mediates microglial synaptic refinement via phosphatidylserine binding. EMBO J 39, e104136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [244].Sapar ML, Ji H, Wang B, Poe AR, Dubey K, Ren X, Ni JQ, Han C (2018) Phosphatidylserine Externalization Results from and Causes Neurite Degeneration in Drosophila. Cell Rep 24, 2273–2286. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [245].Wakatsuki S, Tokunaga S, Shibata M, Araki T (2017) GSK3B-mediated phosphorylation of MCL1 regulates axonal autophagy to promote Wallerian degeneration. J Cell Biol 216, 477–493. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [246].Arispe N, Pollard HB, Rojas E (1993) Giant multilevel cation channels formed by Alzheimer disease amyloid beta-protein [A beta P-(1-40)] in bilayer membranes. Proc Natl Acad Sci U S A 90, 10573–10577. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [247].Ciudad S, Puig E, Botzanowski T, Meigooni M, Arango AS, Do J, Mayzel M, Bayoumi M, Chaignepain S, Maglia G, Cianferani S, Orekhov V, Tajkhorshid E, Bardiaux B, Carulla N (2020) Abeta(1-42) tetramer and octamer structures reveal edge conductivity pores as a mechanism for membrane damage. Nat Commun 11, 3014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [248].Wu J, Blum TB, Farrell DP, DiMaio F, Abrahams JP, Luo J (2021) Cryo-electron Microscopy Imaging of Alzheimer’s Amyloid-beta 42 Oligomer Displayed on a Functionally and Structurally Relevant Scaffold. Angew Chem Int Ed Engl 60, 18680–18687. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [249].Bode DC, Freeley M, Nield J, Palma M, Viles JH (2019) Amyloid-beta oligomers have a profound detergent-like effect on lipid membrane bilayers, imaged by atomic force and electron microscopy. J Biol Chem 294, 7566–7572. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [250].Vander Zanden CM, Wampler L, Bowers I, Watkins EB, Majewski J, Chi EY (2019) Fibrillar and Nonfibrillar Amyloid Beta Structures Drive Two Modes of Membrane-Mediated Toxicity. Langmuir 35, 16024–16036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [251].Flagmeier P, De S, Michaels TCT, Yang X, Dear AJ, Emanuelsson C, Vendruscolo M, Linse S, Klenerman D, Knowles TPJ, Dobson CM (2020) Direct measurement of lipid membrane disruption connects kinetics and toxicity of Abeta42 aggregation. Nat Struct Mol Biol 27, 886–891. [DOI] [PubMed] [Google Scholar]
  • [252].Flagmeier P, De S, Wirthensohn DC, Lee SF, Vincke C, Muyldermans S, Knowles TPJ, Gandhi S, Dobson CM, Klenerman D (2017) Ultrasensitive Measurement of Ca(2+) Influx into Lipid Vesicles Induced by Protein Aggregates. Angew Chem Int Ed Engl 56, 7750–7754. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [253].Sokolov Y, Kozak JA, Kayed R, Chanturiya A, Glabe C, Hall JE (2006) Soluble amyloid oligomers increase bilayer conductance by altering dielectric structure. J Gen Physiol 128, 637–647. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [254].Dong X, Sun Y, Wei G, Nussinov R, Ma B (2017) Binding of protofibrillar Abeta trimers to lipid bilayer surface enhances Abeta structural stability and causes membrane thinning. Phys Chem Chem Phys 19, 27556–27569. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [255].Stevens B, Allen NJ, Vazquez LE, Howell GR, Christopherson KS, Nouri N, Micheva KD, Mehalow AK, Huberman AD, Stafford B, Sher A, Litke AM, Lambris JD, Smith SJ, John SW, Barres BA (2007) The classical complement cascade mediates CNS synapse elimination. Cell 131, 1164–1178. [DOI] [PubMed] [Google Scholar]
  • [256].Schafer DP, Lehrman EK, Kautzman AG, Koyama R, Mardinly AR, Yamasaki R, Ransohoff RM, Greenberg ME, Barres BA, Stevens B (2012) Microglia sculpt postnatal neural circuits in an activity and complement-dependent manner. Neuron 74, 691–705. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [257].Hong S, Beja-Glasser VF, Nfonoyim BM, Frouin A, Li S, Ramakrishnan S, Merry KM, Shi Q, Rosenthal A, Barres BA, Lemere CA, Selkoe DJ, Stevens B (2016) Complement and microglia mediate early synapse loss in Alzheimer mouse models. Science 352, 712–716. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [258].Chung WS, Clarke LE, Wang GX, Stafford BK, Sher A, Chakraborty C, Joung J, Foo LC, Thompson A, Chen C, Smith SJ, Barres BA (2013) Astrocytes mediate synapse elimination through MEGF10 and MERTK pathways. Nature 504, 394–400. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [259].Suzuki J, Denning DP, Imanishi E, Horvitz HR, Nagata S (2013) Xk-related protein 8 and CED-8 promote phosphatidylserine exposure in apoptotic cells. Science 341, 403–406. [DOI] [PubMed] [Google Scholar]
  • [260].Suzuki J, Imanishi E, Nagata S (2014) Exposure of phosphatidylserine by Xk-related protein family members during apoptosis. J Biol Chem 289, 30257–30267. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [261].Segawa K, Kurata S, Yanagihashi Y, Brummelkamp TR, Matsuda F, Nagata S (2014) Caspase-mediated cleavage of phospholipid flippase for apoptotic phosphatidylserine exposure. Science 344, 1164–1168. [DOI] [PubMed] [Google Scholar]
  • [262].Louneva N, Cohen JW, Han LY, Talbot K, Wilson RS, Bennett DA, Trojanowski JQ, Arnold SE (2008) Caspase-3 is enriched in postsynaptic densities and increased in Alzheimer’s disease. Am J Pathol 173, 1488–1495. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [263].Su JH, Zhao M, Anderson AJ, Srinivasan A, Cotman CW (2001) Activated caspase-3 expression in Alzheimer’s and aged control brain: correlation with Alzheimer pathology. Brain Res 898, 350–357. [DOI] [PubMed] [Google Scholar]
  • [264].Gastard MC, Troncoso JC, Koliatsos VE (2003) Caspase activation in the limbic cortex of subjects with early Alzheimer’s disease. Ann Neurol 54, 393–398. [DOI] [PubMed] [Google Scholar]
  • [265].Selznick LA, Holtzman DM, Han BH, Gokden M, Srinivasan AN, Johnson EM Jr, Roth KA (1999) In situ immunodetection of neuronal caspase-3 activation in Alzheimer disease. J Neuropathol Exp Neurol 58, 1020–1026. [DOI] [PubMed] [Google Scholar]
  • [266].Frank MG, Issa NP, Stryker MP (2001) Sleep enhances plasticity in the developing visual cortex. Neuron 30, 275–287. [DOI] [PubMed] [Google Scholar]
  • [267].Li W, Ma L, Yang G, Gan WB (2017) REM sleep selectively prunes and maintains new synapses in development and learning. Nat Neurosci 20, 427–437. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [268].Yang G, Lai CS, Cichon J, Ma L, Li W, Gan WB (2014) Sleep promotes branch-specific formation of dendritic spines after learning. Science 344, 1173–1178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [269].Dumoulin Bridi MC, Aton SJ, Seibt J, Renouard L, Coleman T, Frank MG (2015) Rapid eye movement sleep promotes cortical plasticity in the developing brain. Sci Adv 1, e1500105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [270].Aton SJ, Seibt J, Dumoulin M, Jha SK, Steinmetz N, Coleman T, Naidoo N, Frank MG (2009) Mechanisms of sleep-dependent consolidation of cortical plasticity. Neuron 61, 454–466. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [271].Luo J, Phan TX, Yang Y, Garelick MG, Storm DR (2013) Increases in cAMP, MAPK activity, and CREB phosphorylation during REM sleep: implications for REM sleep and memory consolidation. J Neurosci 33, 6460–6468. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [272].Zhou Y, Lai CSW, Bai Y, Li W, Zhao R, Yang G, Frank MG, Gan WB (2020) REM sleep promotes experience-dependent dendritic spine elimination in the mouse cortex. Nat Commun 11, 4819. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [273].Cohen SI, Linse S, Luheshi LM, Hellstrand E, White DA, Rajah L, Otzen DE, Vendruscolo M, Dobson CM, Knowles TP (2013) Proliferation of amyloid-beta42 aggregates occurs through a secondary nucleation mechanism. Proc Natl Acad Sci U S A 110, 9758–9763. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [274].Scheidt T, Lapinska U, Kumita JR, Whiten DR, Klenerman D, Wilson MR, Cohen SIA, Linse S, Vendruscolo M, Dobson CM, Knowles TPJ, Arosio P (2019) Secondary nucleation and elongation occur at different sites on Alzheimer’s amyloid-beta aggregates. Sci Adv 5, eaau3112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [275].Pandi-Perumal SR, Srinivasan V, Spence DW, Cardinali DP (2007) Role of the melatonin system in the control of sleep: therapeutic implications. CNS Drugs 21, 995–1018. [DOI] [PubMed] [Google Scholar]
  • [276].Ziegler KA, Ahles A, Dueck A, Esfandyari D, Pichler P, Weber K, Kotschi S, Bartelt A, Sinicina I, Graw M, Leonhardt H, Weckbach LT, Massberg S, Schifferer M, Simons M, Hoeher L, Luo J, Erturk A, Schiattarella GG, Sassi Y, Misgeld T, Engelhardt S (2023) Immune-mediated denervation of the pineal gland underlies sleep disturbance in cardiac disease. Science 381, 285–290. [DOI] [PubMed] [Google Scholar]
  • [277].Ouyang M, Hellman K, Abel T, Thomas SA (2004) Adrenergic signaling plays a critical role in the maintenance of waking and in the regulation of REM sleep. J Neurophysiol 92, 2071–2082. [DOI] [PubMed] [Google Scholar]
  • [278].Hunsley MS, Palmiter RD (2004) Altered sleep latency and arousal regulation in mice lacking norepinephrine. Pharmacol Biochem Behav 78, 765–773. [DOI] [PubMed] [Google Scholar]
  • [279].Hayat H, Regev N, Matosevich N, Sales A, Paredes-Rodriguez E, Krom AJ, Bergman L, Li Y, Lavigne M, Kremer EJ, Yizhar O, Pickering AE, Nir Y (2020) Locus coeruleus norepinephrine activity mediates sensory-evoked awakenings from sleep. Sci Adv 6, eaaz4232. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [280].Hablitz LM, Pla V, Giannetto M, Vinitsky HS, Staeger FF, Metcalfe T, Nguyen R, Benrais A, Nedergaard M (2020) Circadian control of brain glymphatic and lymphatic fluid flow. Nat Commun 11, 4411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [281].Iliff JJ, Wang M, Liao Y, Plogg BA, Peng W, Gundersen GA, Benveniste H, Vates GE, Deane R, Goldman SA, Nagelhus EA, Nedergaard M (2012) A paravascular pathway facilitates CSF flow through the brain parenchyma and the clearance of interstitial solutes, including amyloid beta. Sci Transl Med 4, 147ra111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [282].Porkka-Heiskanen T, Strecker RE, Thakkar M, Bjorkum AA, Greene RW, McCarley RW (1997) Adenosine: a mediator of the sleep-inducing effects of prolonged wakefulness. Science 276, 1265–1268. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [283].Peng W, Wu Z, Song K, Zhang S, Li Y, Xu M (2020) Regulation of sleep homeostasis mediator adenosine by basal forebrain glutamatergic neurons. Science 369. [DOI] [PubMed] [Google Scholar]
  • [284].Halassa MM, Florian C, Fellin T, Munoz JR, Lee SY, Abel T, Haydon PG, Frank MG (2009) Astrocytic modulation of sleep homeostasis and cognitive consequences of sleep loss. Neuron 61, 213–219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [285].Lee MR, Ruby CL, Hinton DJ, Choi S, Adams CA, Young Kang N, Choi DS (2013) Striatal adenosine signaling regulates EAAT2 and astrocytic AQP4 expression and alcohol drinking in mice. Neuropsychopharmacology 38, 437–445. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [286].Shi Z, Zhang W, Lu Y, Lu Y, Xu L, Fang Q, Wu M, Jia M, Wang Y, Dong L, Yan X, Yang S, Yuan F (2017) Aquaporin 4-Mediated Glutamate-Induced Astrocyte Swelling Is Partially Mediated through Metabotropic Glutamate Receptor 5 Activation. Front Cell Neurosci 11, 116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [287].Mestre H, Hablitz LM, Xavier AL, Feng W, Zou W, Pu T, Monai H, Murlidharan G, Castellanos Rivera RM, Simon MJ, Pike MM, Pla V, Du T, Kress BT, Wang X, Plog BA, Thrane AS, Lundgaard I, Abe Y, Yasui M, Thomas JH, Xiao M, Hirase H, Asokan A, Iliff JJ, Nedergaard M (2018) Aquaporin-4-dependent glymphatic solute transport in the rodent brain. Elife 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [288].Gomolka RS, Hablitz LM, Mestre H, Giannetto M, Du T, Hauglund NL, Xie L, Peng W, Martinez PM, Nedergaard M, Mori Y (2023) Loss of aquaporin-4 results in glymphatic system dysfunction via brain-wide interstitial fluid stagnation. Elife 12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [289].Inostroza M, Born J (2013) Sleep for preserving and transforming episodic memory. Annu Rev Neurosci 36, 79–102. [DOI] [PubMed] [Google Scholar]
  • [290].Krause AJ, Simon EB, Mander BA, Greer SM, Saletin JM, Goldstein-Piekarski AN, Walker MP (2017) The sleep-deprived human brain. Nat Rev Neurosci 18, 404–418. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [291].Tononi G, Cirelli C (2014) Sleep and the price of plasticity: from synaptic and cellular homeostasis to memory consolidation and integration. Neuron 81, 12–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [292].de Vivo L, Bellesi M, Marshall W, Bushong EA, Ellisman MH, Tononi G, Cirelli C (2017) Ultrastructural evidence for synaptic scaling across the wake/sleep cycle. Science 355, 507–510. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [293].Miyamoto D, Marshall W, Tononi G, Cirelli C (2021) Net decrease in spine-surface GluA1-containing AMPA receptors after post-learning sleep in the adult mouse cortex. Nat Commun 12, 2881. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [294].Maret S, Faraguna U, Nelson AB, Cirelli C, Tononi G (2011) Sleep and waking modulate spine turnover in the adolescent mouse cortex. Nat Neurosci 14, 1418–1420. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [295].Cary BA, Turrigiano GG (2021) Stability of neocortical synapses across sleep and wake states during the critical period in rats. Elife 10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [296].Park JM, Hu JH, Milshteyn A, Zhang PW, Moore CG, Park S, Datko MC, Domingo RD, Reyes CM, Wang XJ, Etzkorn FA, Xiao B, Szumlinski KK, Kern D, Linden DJ, Worley PF (2013) A prolyl-isomerase mediates dopamine-dependent plasticity and cocaine motor sensitization. Cell 154, 637–650. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [297].Okada D, Ozawa F, Inokuchi K (2009) Input-specific spine entry of soma-derived Vesl-1S protein conforms to synaptic tagging. Science 324, 904–909. [DOI] [PubMed] [Google Scholar]
  • [298].Chokshi V, Gao M, Grier BD, Owens A, Wang H, Worley PF, Lee HK (2019) Input-Specific Metaplasticity in the Visual Cortex Requires Homer1a-Mediated mGluR5 Signaling. Neuron 104, 736–748 e736. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [299].Kato A, Fukazawa Y, Ozawa F, Inokuchi K, Sugiyama H (2003) Activation of ERK cascade promotes accumulation of Vesl-1S/Homer-1a immunoreactivity at synapses. Brain Res Mol Brain Res 118, 33–44. [DOI] [PubMed] [Google Scholar]
  • [300].Schneider B, Mutel V, Pietri M, Ermonval M, Mouillet-Richard S, Kellermann O (2003) NADPH oxidase and extracellular regulated kinases 1/2 are targets of prion protein signaling in neuronal and nonneuronal cells. Proc Natl Acad Sci U S A 100, 13326–13331. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [301].Toni M, Spisni E, Griffoni C, Santi S, Riccio M, Lenaz P, Tomasi V (2006) Cellular prion protein and caveolin-1 interaction in a neuronal cell line precedes Fyn/Erk 1/2 signal transduction. J Biomed Biotechnol 2006, 69469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [302].Caetano FA, Lopes MH, Hajj GN, Machado CF, Pinto Arantes C, Magalhaes AC, Vieira Mde P, Americo TA, Massensini AR, Priola SA, Vorberg I, Gomez MV, Linden R, Prado VF, Martins VR, Prado MA (2008) Endocytosis of prion protein is required for ERK1/2 signaling induced by stress-inducible protein 1. J Neurosci 28, 6691–6702. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [303].Chiarini LB, Freitas AR, Zanata SM, Brentani RR, Martins VR, Linden R (2002) Cellular prion protein transduces neuroprotective signals. EMBO J 21, 3317–3326. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [304].Krapivinsky G, Krapivinsky L, Manasian Y, Ivanov A, Tyzio R, Pellegrino C, Ben-Ari Y, Clapham DE, Medina I (2003) The NMDA receptor is coupled to the ERK pathway by a direct interaction between NR2B and RasGRF1. Neuron 40, 775–784. [DOI] [PubMed] [Google Scholar]
  • [305].Li S, Tian X, Hartley DM, Feig LA (2006) Distinct roles for Ras-guanine nucleotide-releasing factor 1 (Ras-GRF1) and Ras-GRF2 in the induction of long-term potentiation and long-term depression. J Neurosci 26, 1721–1729. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [306].Wang Y, Shi Z, Zhang Y, Yan J, Yu W, Chen L (2020) Oligomer beta-amyloid Induces Hyperactivation of Ras to Impede NMDA Receptor-Dependent Long-Term Potentiation in Hippocampal CA1 of Mice. Front Pharmacol 11, 595360. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES