Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2025 Oct 1.
Published in final edited form as: J Alzheimers Dis. 2025 Jul 1;106(2):391–409. doi: 10.1177/13872877251342273

The interaction between neurotransmitter receptor activity and amyloid-β pathology in Alzheimer’s disease

Yuhan Nong 1, Jung Soo Kim 1, Litian Jia 1, Ottavio Arancio 2, Qi Wang 1,3,*
PMCID: PMC12283230  NIHMSID: NIHMS2089756  PMID: 40388923

Abstract

The accumulation of amyloid-β (Aβ) peptides is a hallmark of Alzheimer’s disease (AD). Central to AD pathology is the production of Aβ peptides through the amyloidogenic processing of amyloid precursor protein (APP) by β-secretase (BACE-1) and γ-secretase. Recent studies have shifted focus from Aβ plaque deposits to the more toxic soluble Aβ oligomers. One significant way in which Aβ peptides impair neuronal information processing is by influencing neurotransmitter receptor function. These receptors, including adrenergic, acetylcholine, dopamine, 5-HT, glutamate, and gamma-aminobutyric acid (GABA) receptors, play a crucial role in regulating synaptic transmission, which underlies perceptual and cognitive functions. This review explores how Aβ interacts with these key neurotransmitter receptors and how these interactions contribute to neural dysfunction in AD. Moreover, we examine how agonists and antagonists of these receptors influence Aβ pathology, offering new perspectives on potential therapeutic strategies to curb AD progression effectively and improve patients’ quality of life.

Keywords: Alzheimer’s disease, Amyloid-β, Norepinephrine, Acetylcholine, Dopamine, 5HT, Glutamate, GABA, neurotransmitter

I. Introduction

Alzheimer’s disease (AD) is one of the leading causes of dementia in the world. The pathology of AD includes the aggregation of amyloid-β (Aβ) proteins, the formation of insoluble plaques, and the production of neurofibrillary tangles resulting from hyperphosphorylated Tau protein aggregation. Most amyloid precursor protein (APP) is processed through the non-amyloidogenic pathway, where α-secretase cleaves APP to produce soluble APPα (sAPPα), followed by γ-secretase cleavage, preventing the formation of intact Aβ. In amyloidogenic pathway, sequential proteolytic cleavage of the APP by β-secretase (ß-site APP-cleaving enzyme 1, BACE-1) and γ-secretase, generates Aß peptides ranging in length from 38 to 43 residues.13 Among the various isoforms of Aβ, Aβ42 and Aβ40 are the most commonly observed in human AD patients. Aβ42 is more prone to aggregation, while Aβ40 is less likely to form aggregates.4 Therefore, the Aβ42/Aβ40 ratio serves as a marker for early AD progression. Familial AD is often linked to specific mutations in genes encoding APP and the catalytic subunit of γ-secretase, known as presenilin.57 These mutations tend to facilitate the amyloidogenic processing of APP, thereby increasing Aβ production. While amyloid plaque was traditionally regarded as neurotoxic, recent studies have underscored the toxicity of Aβ oligomers,8, 9 including their association with long-term potentiation (LTP) impairment,10 synapse loss,11 and cognitive dysfunction.12

Within the complex context of brain function, neurotransmitter receptors serve as important mediators of neuronal network dynamics underlying neural information processing. These receptors, including adrenergic receptors (ARs), acetylcholine receptors (AchRs), dopamine receptors (DRs), 5-HT receptors (5-HTRs), glutamate receptors (GluRs) and gamma-aminobutyric acid receptors (GABARs) are the key receptors in regulating synaptic transmission and further perceptual and cognitive processes.1317 Aβ has been shown to alter neuronal signaling by interacting with various neurotransmitter receptors (Figure 1).18, 19 Dysregulation or impairment of these receptors in the context of AD can influence neural circuit dynamics and thus exacerbate perceptual and cognitive dysfunction. Despite the continuous effort towards the treatment of AD, little therapeutic progress has been made over the past two decades. Exploring the interplay between Aβ pathology and the activity of these common neurotransmitter receptors would be critical in uncovering the mechanisms underlying AD pathogenesis and identifying new therapeutic strategies.

Figure 1.

Figure 1.

The production of amyloid-β (Aβ) peptides and their interaction with neurotransmitter receptor-mediated signaling pathways. Illustration was created in https://biorender.com

In this review, we focus on elucidating the role of neurotransmitter receptors in shaping the outcomes of AD treatment. By exploring the evidence and mechanisms through which the activity of neurotransmitter receptors interacts with Aβ pathology, we aim to provide insights that could potentially inform the development of therapeutic interventions for mitigating the impact of AD on brain functions and quality of life.

II. Interaction between adrenergic receptor activity and Aß pathology

Adrenergic receptors are a class of G protein-coupled receptors that are targets of norepinephrine. These receptors play essential roles in the regulation of a variety of brain functions.2022 They are broadly classified into α1, α2 and β subtypes. In the CNS, α2 and β2 adrenergic receptors are the most common. The α2 receptors inhibit adenylyl cyclase activity through Gi proteins, while β receptors stimulate it via Gs proteins. Meanwhile, α1 receptors activate Phospholipase C (PLC) through Gq proteins.23 Studies have demonstrated that AD is related to the change of locus coeruleus – norepinephrine (LC-NE) system.2426 For instance, AD mouse models exhibited age-related loss of LC neurons,27 and this loss coincided with increasing ß adrenergic receptor activity.28 Another example is by increasing ß adrenergic activity, researchers preserved Aß-induced LTP impairment in AD mouse models.29 Thus, it is important to elucidate different types of adrenergic receptors and their interplay with Aß.

2.1. ß2 adrenergic receptors

The β2 adrenergic receptor has been the most studied among all the adrenergic receptor subtypes. Soluble Aß can directly bind to the ß2 adrenergic receptor and trigger a cascade of downstream reactions, including activation of Protein Kinase A (PKA) signaling for GluR1 phosphorylation and enhancement of AMPAR-mediated excitatory postsynaptic current (EPSCs).30, 31 The majority of studies on enhancing ß2 adrenergic receptor activity concluded that the activation of β2 adrenergic receptors plays a protective role against Aβ toxicity. Specifically, isoproterenol treatment and enriched environments, both of which stimulate β2 adrenergic receptors, have been shown to counteract Aβ-induced hippocampal impairments by activating cyclic AMP (cAMP)-PKA pathway.32 Moreover, activation of the β2 adrenergic receptor by clenbuterol not only reduced Aβ plaque accumulation by modulating APP metabolism on molecular level, but also promoted hippocampal neurogenesis and memory function.33, 34 These protective effects extend to the epigenetic level, with procaterol, another β2 adrenergic receptor agonist, inhibiting Aβ-induced synaptotoxicity through regulating histone acetylation.35 From a translational perspective, human subjects receiving β2 adrenergic receptor agonists exhibited a reduced risk of developing AD, emphasizing the therapeutic potential of these compounds.36 In contrast, some research suggested a potential adverse effect of ß2 adrenergic receptor agonists. Clenbuterol and isoproterenol have been implicated in accelerating hippocampal and cerebral amyloid production, likely due to the enhanced γ-secretase activity.37, 38

The inhibition of ß2 adrenergic receptors, for example by its antagonist ICI118551, has also yielded contrasting findings. Studies have reported that ICI118551 can attenuate acute stress-induced Aß production37 and even reduce Aß plaque formation with chronic treatment,38 suggesting potential neuroprotective effects. In contrast, other investigations have revealed that the antagonist can elevate Aß levels through increasing amyloidogenic APP processing30, 39 and induce cognitive deficits in AD model mice by inhibiting dendritic ramification.39 These discrepancies likely arise from a variety of factors, including differences in AD models used, the dosage and duration of pharmacological treatment, and the stage of disease progression. Addressing these variables through standardized protocols and comprehensive studies will be essential for uncovering the precise role and therapeutic potential of β2 adrenergic receptors in AD.

2.2. α2 receptors

Research focusing on the interface of Aß and α2 adrenergic receptor has revealed surprising molecular interactions relevant to AD progression. Specifically, Aß oligomers can bind to the α2 adrenergic receptor with nanomolar affinity, redirecting NE signaling, triggering the glycogen synthase kinase-3 ß (GSK3β) cascade and resulting in Tau hyperphosphorylation.40 Moreover, APP also directly interacts with the α2A adrenergic receptor subtype, decreasing receptor internalization and potentially modulating NE signaling.41 As pharmacological interventions, activation of the α2A adrenergic receptor subtype by clonidine has been shown to exacerbate Aß production by disrupting the interaction between APP and Sorting-related receptor with A repeat (SorLA).42 Conversely, inhibiting the α2 adrenergic receptor could reduce Aß generation and rescue Aß-induced cognition dysfunction.42, 43

Beyond ß2 and α2 adrenergic receptors, researchers also explored the effect of other subtypes of adrenergic receptor on AD progression related to Aß. For example, researchers found that treatment of CL316243, a ß3 adrenergic receptor agonist, effectively rescued Aß-induced memory dysfunction and reduced Aß42/Aß40 ratio.44, 45 Furthermore, by inhibiting α1 adrenergic receptor in AD model mice, BACE1 expression and GSK3ß phosphorylation were reduced, which in turn resulted in less Aß production and better behavior performance.46

III. Interaction between cholinergic receptor activity and Aß pathology

AchRs are divided into two types: muscarinic (mAchRs), which are G-protein coupled receptors, and nicotinic (nAchRs), which are ionotropic receptors. Muscarinic receptors have five subtypes (M1-M5),47 among which the M1 subtype is prominently expressed in the central nervous system and is significantly associated with AD.48 nAchRs are found in various subtypes, each with unique properties and distinct distributions within the brain.49 Research has demonstrated Aß effects on both nAchRs and mAchRs.5052

3.1. α7-nAch receptors

The α7 nicotinic acetylcholine receptor is a homomeric receptor composed solely of five identical α7 subunits.53 This receptor is known for its high calcium permeability, which distinguishes it from many other nAChR subtypes. It is widely distributed in the central nervous system and plays a role in cognitive function, learning, memory, and synaptic plasticity. Research has shown Aβ oligomers could bind to the orthosteric binding site of α7-nAChR with high affinity,54, 55 and this binding induces concentration-dependent conformational changes on α7nAChR.56 Consistent with results from several studies that have shown the effects of Aβ on neuronal activity and synaptic function, Aß itself directly influenced α7-nAChR’s function by acting as a negative modulator to reduce their activation duration,56 indicating that Aß can functionally act as an α7nAchR antagonist.57, 58 While Aβ exposure has been shown to lead to unpredictable alterations in membrane potential and decrease in excitatory postsynaptic potentials (EPSPs) through L-type calcium channels,59 it also resulted in post-translational and functional upregulation of α7-nAChRs60, 61 and suppression of nAchR agonist-induced excitation.62, 63 Interestingly, early Aβ-induced neuron hyperactivity, mediated by α7-nAChR, usually followed by synaptic inhibition.64 These results highlighted a complex effect of Aß on α7-nAchR. Moreover, the interaction of Aβ with α7-nAChRs can alter the dynamic properties of neuronal networks in amyloid overproducing mice.65 The role of familial AD-associated Arctic Aβ has also been investigated, revealing its ability to bind to the nAChR α7 subunit and inhibit the calcium ion response and ERK1/2 activation.66

Interestingly, both activation and inhibition of α7-nAChR yielded beneficial outcomes regarding Aß toxicity. Agonists of α7-nAChR, such as epibatidine, SSR180711, and A-582941, have been shown to protect receptor loss from Aß42 toxicity,54 reverse Aβ-induced synaptic transmission deficit,67 and enhance cognitive functions or induce neuroprotective effects in WT mice or mouse models of AD.68, 69 These effects may result from the ability of α7-nAChR agonists to disrupt the interaction between Aß and nAChR.70 Conversely, α7-nAChR antagonists, like methyllycaconitine, have also demonstrated inhibitory effects on certain Aβ-induced toxicities, highlighting the complex role of α7-nAChR in AD pathology. For example, methyllycaconitine can prevent and inverse Aß binding to α7nAchR and prevent Aß induced neuronal hyperexcitation.55, 61 Also, α7-nAChR antagonist cotreated in cell culture with Aß42 could diminish Aß42 associated mitochondrial dysfunction.71 Additionally, genetic approaches, such as the knockout of the α7-nAChR gene, have been shown to trigger age-dependent AD-like pathology.72 However, some research suggested that this deletion may result in rescuing cognitive deficits and synaptic pathology in certain AD models.61, 73

3.2. α7β2 nAch receptors

The α7β2 nAchR subtype represents a more recently identified and less understood configuration, comprising both α7 and β2 subunits.74 This heteromeric assembly diversifies the functional and pharmacological properties of the receptors to other nAchRs.75 For example, their choline-induced current showed smaller amplitude and a longer duration compared to homomeric α7-nAchR.75 Researchers also found that α7β2-nAchR exhibits greater sensitivity to Aβ42 oligomers compared to α7-nAchR, as its function can be blocked by lower nanomolar concentrations of Aβ42 oligomers.76, 77 This heightened sensitivity may indicate a unique role for α7β2 nAchR in AD pathogenesis. Activation of α7β2-nAChR receptors by Aβ42 oligomers also triggered hyperexcitation and degeneration of basal forebrain cholinergic neurons.78 At molecular level, Aβ42 oligomers also preferentially extend the open-dwell times of α7β2-nAChR, likely contributing to cognitive decline in AD.78 In addition, APP/PS1 transgenic mice lacking α7β2-nAChR displayed improved spatial reference memory compared to normal APP/PS1 transgenic mice, further emphasizing the significance of this receptor subtype in AD pathology.78

3.3. α4β2 nAch receptors

The α4β2 subtype is one of the most abundant nicotinic receptors in the brain. Compared to the α7-nAchR, it has slower activation and desensitization kinetics and lower calcium permeability.79, 80 The α4β2-nAchR also plays a critical role in modulating synaptic plasticity and cognitive processes. Essential insights about α4β2-nAchR in AD come from studies that showed Aß reducing the expression of α4β2-nAchR in cell culture.81 Epibatidine is a potent agonist of α4β2-nAchR and a less potent agonist of α7-nAchR.82 Aβ’s ability to suppress epibatidine-induced currents in rat hippocampal CA1 pyramidal neurons demonstrated its negative impact on the α4β2- and α7-nAchR subtype.63 Additional findings revealed that inhibition of Aß40 on AchR-evoked dopamine release is partly mediated by the extracellular interaction between α4β2-nAchR and Aß40 molecule.83 The ability of Aß on selectively targeting α7- and α4β2-nAChRs attributed to its interaction with arginine 208 and glutamate 211 on the α7 & α4 subunits84. Moreover, coactivation of α7- and α4β2-nAChRs reversed AMPAR dysfunction and LTP disruption induced by Aβ.84

3.4. mAch receptors

mAChRs are a class of G protein-coupled receptors that respond to the neurotransmitter acetylcholine. Unlike nAChRs, which are ion channels, mAChRs influence cells through a variety of signal transduction pathways.85 There are five known subtypes of mAChRs, designated M1 through M5, each with distinct functions and pharmacological profiles. A study showed that M1 receptors significantly modulate AD-like pathology in 3xTg-AD transgenic mice, with M1 mAChR agonist AF267B leading to an improvement in memory performance and a reduction in amyloid and Tau pathology.86 This study also found that dicyclomine, an M1 antagonist, yielded opposite results, thereby emphasizing the potential therapeutic importance of M1 mAChR.86 The interaction of Aß and mAChR extends to synaptic function as well. It has been demonstrated that Aß-induced EPSC reduction could be mitigated by atropine, a mAChR antagonist, and calcicludine, a calcium channel antagonist.51 This finding also indicated that the mAChR plays a more fundamental role than the nAChR in this context since nAchR antagonist has no such effect. Complementing this observation, another study showed that the enhanced activation of intracellular signaling enzymes Protein Kinase C (PKC) and CaMKII by Aß could be inhibited by oxo-M, an mAChR agonist.87 However, nAchR agonist had no such effect.87 Further complicating the Aß-mAChR interaction, a study suggested that Aß oligomers interfere with the functionality of the M1 mAChR by altering its interaction with G-proteins88 or modulate M5 mAchR signal transduction intracellularly.83 Together, it is clear that the mechanisms through which Aß and mAChR interact in AD are complicated, and future research is needed to further elucidate these complex interactions.

IV. Interaction between dopaminergic receptor activity and Aß pathology

In addition to adrenergic receptors, dopaminergic receptors represent another major type of catecholamine receptors. Dopamine receptors are a class of GPCRs and consist of five main subtypes, labeled D1 through D5, which are divided into two main classes based on their pharmacological properties and effects: the D1-like receptors (D1 and D5) and the D2-like receptors (D2, D3, and D4).89 In the CNS, the most prominent subtypes are D1 and D2 receptors. D1 receptors, primarily excitatory, are coupled to Gs proteins, which increase the cAMP and thus promoting cellular signaling pathways that modulate motor control, cognition, and reward systems. In contrast, D2 receptors are inhibitory and coupled to Gi proteins, which reduce cAMP and modulate neuronal excitability. Because D2 receptors are critically involved in regulating motor functions, mood, and motivation, they serve as essential pharmacological targets in the treatment of disorders like Parkinson’s disease and schizophrenia.90, 91 In comparison to PD and schizophrenia, research on dopamine receptor involvement in AD is relatively limited. However, altered dopamine receptor levels have been observed in AD patients.9294 PET imaging studies have demonstrated reduced D2 receptor binding availability in AD brains.94 Immunohistochemical analyses revealed significantly decreased expression of cortical D1, D3, and D4 receptors, while D5 receptor expression was elevated.93

4.1. Dopaminergic D1 receptors

Recent studies have begun to explore the effects of dopamine receptor modulation on amyloid pathology. For instance, D1 receptor agonists such as SKF38393 and A-68930 have shown promise in mitigating cognitive deficits induced by intracerebroventricular (icv)-injected Aß.95, 96 The underlying mechanisms remain under investigation, with researchers identifying different pathways of action. SKF38393 has been found to increase cAMP response element binding protein (CREB) phosphorylation, subsequently enhancing Brain-Derived Neurotrophic Factor (BDNF) and B-cell Lymphoma 2 (Bcl-2) expression and reducing BACE1 activity.96 Meanwhile, A-68930 appeared to alleviate Aβ-induced neuroinflammation via an AMPK/autophagy pathway, promoting NLR family pyrin domain containing 3 (NLRP3) inflammasome degradation and reducing IL-1β and IL-18 levels.95 Conversely, the D1 receptor antagonist SCH23390 demonstrated opposite effects to SKF38393, further underscoring the therapeutic potential of D1 receptor agonists.96

4.2. Dopaminergic D2 receptors

In studying the effect of dopaminergic D2 receptor activity in AD, bromocriptine, a D2 receptor agonist, demonstrated protective effects against cognitive impairment induced by icv-injected Aß.97 Mechanistic studies in both in vivo and in vitro models revealed that bromocriptine, through dopaminergic D2 receptor activation, recruited protein phosphatase 2A (PP2A) and c-Jun N-terminal kinase (JNK) via β-arrestin 2. This action inhibited JNK-mediated transcription of proinflammatory cytokines and prevented NLRP3 inflammasome activation in microglia. Rotigotine, another D2/D3 receptor agonist, has been shown to increase cortical excitability and restore central cholinergic transmission in AD patients.98

V. Interaction between 5-HT receptor activity and Aß pathology

The 5-HT (serotonin) receptors are a diverse group of receptors that mediate the effects of serotonin across both the central and peripheral nervous systems. They are classified into seven families, from 5-HT1 to 5-HT7, with most subtypes being GPCRs that modulate intracellular signaling pathways.99 An exception is the 5-HT3 receptor, which is a ligand-gated ion channel responsible for fast excitatory neurotransmission. The 5-HT1 family, including subtypes like 5-HT1A and 5-HT1B, is primarily inhibitory, reducing cAMP levels and decreasing neuronal excitability. On the other hand, families such as 5-HT2, 5-HT4, and 5-HT6 are excitatory, with 5-HT2 increasing intracellular calcium through Gq signaling and 5-HT4 and 5-HT6 stimulating cAMP production via Gs signaling.100 Each subtype shows a distinct pattern of expression across brain regions and contributes to a wide range of functions, including mood regulation, cognition, appetite, and circadian rhythms. For instance, 5-HT1AR is highly expressed in the midbrain, limbic system (especially the hippocampus), and cortex, while 5-HT2AR is predominantly located in the cortex, particularly in high-level associative.101 Research has shown that Aß affects the serotonergic system, disrupting normal 5-HT receptor signaling.102 The interaction between Aß and 5-HT1A, 5-HT2A, 5-HT2B, 5-HT4, and 5-HT6 receptors has been the most extensively studied, revealing important insights into their roles in neurodegenerative processes.

5.1. 5-HT1A receptors

The 5-HT1A receptor, part of the 5-HT1 receptor family, is highly expressed in brain regions such as the hippocampus and amygdala, which are crucial for emotional processing and cognitive functions. It is primarily an inhibitory GPCR that reduces cAMP production by inhibiting adenylate cyclase, therefore decreasing neuronal excitability. PET imaging studies have shown altered 5-HT1A receptor expression during different stages of AD, with some studies reporting a reduction in 5-HT1A density,103, 104 while others have observed an upregulation.105, 106 The interaction between Aß and 5-HT1A receptors presented complex effects on neuronal function and cognitive outcomes in AD models. Aß40 and Aß42 differentially influenced 5-HT1AR expression: Aß40 induced receptor overexpression, possibly as a protective mechanism, whereas Aß42 caused neuronal lesions without affecting receptor levels.107 In models of memory loss induced by streptozotocin (STZ), which mimic memory impairments in AD, the 5-HT1AR antagonist NAD-299 mitigated memory deficits, reduced oxidative stress, and decreased neuronal loss.108, 109 Additionally, the 5-HT1AR antagonist WAY100635 reduced neuroinflammation and improved cognitive performance in Aß42-injected mice, possibly through the NF-κB pathway.110

5.2. 5-HT2 receptors

Research has shown both AD mouse model and human patients exhibit a loss of 5-HT2A receptors in various brain regions, including the hippocampus, medial prefrontal cortex (mPFC) and cerebral cortex.111115 Studies using PET imaging in AD patients have revealed a reduction in cortical 5-HT2AR binding, independent of serotonergic neuron loss. This finding suggested that 5-HT2AR loss may be an early feature of AD.111 Injecting Aß42 into the hippocampus led to reductions in BDNF, memory deficits, and a loss of hippocampal 5-HT2AR.114, 115 Both 5-HT2AR agonists and antagonists showed therapeutic potential in AD treatments. In a STZ-induced rat model of memory loss, the 5-HT2AR agonist TCB-2 has been shown to alleviate memory deficits, reduce oxidative stress, and mitigate neuronal loss, suggesting a neuroprotective effect.108, 109 Moreover, Desloratadine (DLT), a selective 5-HT2AR antagonist, reduced Aß plaque deposition in AD model mice by facilitating microglial phagocytosis of Aß.116 Unlike the 5-HT2AR, pioneering research indicated that patients with sporadic AD exhibit elevated expression of the 5-HT2BR in the cortex.117 Additionally, there was an increase in 5-HT2BR mRNA expression associated with Aß accumulation.118 The selective 5-HT2BR antagonist, MW701, has demonstrated promising results in mitigating Aß42-induced impairments in LTP and memory deficits.117

5.3. 5-HT4 receptors

The 5-HT4 receptor is another excitatory serotonin receptor. It activates the cAMP-PKA pathway, increasing neuronal excitability. Predominantly expressed in the hippocampus, 5-HT4 receptors are widely recognized for their involvement in learning and memory. Treatment with the 5-HT4R agonist RS-67333, both short-term (two weeks) and long-term (four months), improved memory in AD model mice while reducing Aß load and neuroinflammation.119121 Moreover, treatment with both RS-67333 and Usmarapride (a 5-HT4R partial agonist) increased soluble APPα, indicating the possible involvement of α-secretase activation.121, 122 Research has further suggested that 5-HT4R agonists modulate APP processing to favor the non-amyloidogenic pathway.123125 Activation of α-secretase may occur through the ERK signaling pathway via cAMP and PKA signaling, which activates MEK and ERK, enhancing α-secretase ADAM10 activity and reducing Aß levels.123, 125 Evidence also suggested that 5-HT4R may activate α-secretase through a G-protein and Src-dependent activation of PLC, bypassing cAMP and PKA signaling.124 These findings highlighted the complexity of GPCR signaling in APP processing and Aß metabolism.

5.4. 5-HT6 receptors

The 5-HT6 receptor, primarily expressed in the hippocampus, is important in mediating learning and memory. Treatment with 5-HT6R antagonists, such as AVN-211 and SB-25858, has shown positive effects in attenuating Aß-induced memory loss,120, 126128 likely by regulating the morphology and function of neuronal primary cilia.129 There is also evidence that 5-HT6R agonists can decrease amyloid pathology and prevent memory loss.123, 130 One potential mechanism is that 5-HT6R activation would increase α-secretase activation through the PKA-ERK pathway. However, further research is needed to further clarify the mechanisms by which 5-HT6R modulates amyloid pathology.

Research on the interaction between Aß and other types of 5-HT receptors is limited, but there is potential for discovering novel therapeutic targets for AD. For instance, the 5-HT7R agonist AS19 has shown promising effects in reducing Aß plaque deposition, preventing neuronal apoptosis, and improving memory performance in rat AD models.131, 132

VI. Interaction between Glutamate receptors and Aß pathology

Glutamate is the primary excitatory neurotransmitter in the central nervous system. Given the importance of the glutamate system in memory formation, the interaction between Aß and glutamate receptors has been heavily studied. Glutamate receptors are broadly categorized into ionotropic and metabotropic types. The ionotropic receptors include NMDA (N-methyl-D-aspartate) and AMPA (α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid) receptors, which are directly involved in fast synaptic transmission. Metabotropic glutamate receptors (mGluRs) are G protein-coupled receptors that modulate neuronal and synaptic function through secondary messenger systems. It can be further divided into 3 groups. Group I mGluRs, including mGluR1 and mGluR5, activate phosphoinositide hydrolysis, leading to increased intracellular calcium levels and activation of PKC. Group II mGluRs, such as mGluR2 and mGluR3, and Group III mGluRs, which include mGluR4, mGluR6, and mGluR7, inhibit adenylate cyclase activity, resulting in a reduction of cAMP levels.133 These distinct signaling mechanisms allow mGluRs to play diverse roles in modulating neuronal excitability and synaptic plasticity.

6.1. NMDA receptors

NMDARs are distinguished by their voltage-dependent activation, requiring both glutamate binding and membrane depolarization to relieve Mg2+ block of the ion channel. This makes them highly sensitive to the synaptic activity and capable of detecting coincident pre- and postsynaptic activity, fundamental for the synaptic plasticity processes like LTP. The relationship between Aß and NMDAR is most studied among all glutamate receptors. Aß was shown to decrease the surface expression of NMDAR.134, 135 More specifically, Aß oligomers colocalized with GluN1 and GluN2B, further leading to the loss of GluN2B subunit.136, 137 and a shift in NMDAR composition from GluN2B to GluN2A.138 NMDAR played an important role in Aß-induced spine loss and LTP impairment.139142 One possible reason for Aß oligomers-induced spine loss could be their role in reducing calcium influx into active spine through NMDAR,143 or it could be the hyperactivation of caspase-3.144 All the subunits, GluN1, GluN2A and GluN2B, were required for Aß-induced spine loss.135 As for LTP impairment, Aß has been shown to inhibit LTP by enhancing extrasynaptic NMDAR-mediated function, but not via synaptic NMDAR.140, 141 Among all NMDAR subunits, GluN2B is particularly significant in Aß-related pathology. Aß oligomers mainly targeted hippocampal extrasynaptic NMDAR containing GluN2B141 and resulted in loss of GluN2B response.138 In addition, the dephosphorylation of GluN2B at Tyr1472 was found to correlate with Aß-mediated NMDAR endocytosis.145

Some research indicated NMDAR activation can increase the activity of α-secretase and thus inhibit amyloid pathogenesis.146 However, a different viewpoint claims that after separating the functions of extrasynaptic and synaptic NMDAR, extrasynaptic NMDAR activation can shift APP isoform from APP695 to Kunitz protease inhibitory domain (KPI) containing APP (KPI-APP).147 KPI-APP has a higher amyloidogenic potential, meaning KPI-APP could inhibit α-secretase pathway and enhance ß-secretase processing.148 In parallel, the calcium pathway played an important role in Aß-NMDAR interaction. Studies suggested that Aβ oligomers directly reduce NMDAR-mediated calcium influx, particularly in highly active synapses.134, 143, 149 Furthermore, the dose of Aß needed to block calcium entry for NMDAR is much lower compared to the dose needed to induce NMDAR endocytosis.149 This result indicated a new early-stage impairment of Aß on NMDAR metabolism. However, other evidence suggested that instead of inhibiting calcium entry, Aβ oligomers may enhance calcium influx through the activation of NMDAR, ultimately leading to mitochondrial dysfunction and neuronal loss.150

Inhibition of NMDAR for AD treatment has already been successful on the bedside. Memantine, a non-competitive NMDAR antagonist, has been approved by FDA for AD treatment.151, 152 One potential mechanism of action of memantine therapy might be its ability to block extrasynaptic NMDAR-mediated currents without affecting the normal synaptic NMDAR-mediated currents.153 Studies also found memantine inhibited NMDAR-mediated KPI-APP production, hence inhibiting Aß generation, in a dose-dependent manner.147 Chronic treatment of memantine not only reduced Aß oligomers and prevented Aß-induced LTP impairment,154 but also reduced neuronal loss and prevented memory dysfunction.155, 156 Beside memantine, AP5, another broad NMDAR antagonist, also showed similar effects on preventing Aß-induced LTP impairment157 and calcium level imbalance.158 On the contrary, 3-(2-carboxypiperazin-4yl) propyl-1-phosphonic acid (CPP), also a NMDAR antagonist, increased Aß load in the brain and triggered spine loss.143, 159 People has now been focusing on the manipulation of GluN2B subunit. Ifenprodil and Ro 25–6981, both GluN2B antagonist, showed to prevent Aß oligomer-induced deficit in LTP and NMDAR impairment.149, 160, 161 These findings indicated the significance of GluN2B in Aß pathological impact.

6.2. AMPA receptors

AMPA receptors are the main mediators of fast excitatory synaptic transmission inside the brain. Their rapid activation by glutamate allows for quick changes in membrane potential via the influx of sodium. Research has shown Aß decrease the density of AMPAR in the cortex.162 One possibility is that Aß induced AMPAR ubiquitination via increasing AMPAR E3 ligase Nedd4 and decreasing AMPAR deubiquitinase USP46 expression.163 This process depended on the presence of GluR1 subunit, whose synaptic expression was decreased with the presence of early Aß pathology,164, 165 but the detail mechanism has been controversial. Some argued that it is due to GluR1 phosphorylation, since GluR1 ubiquitination-deficient could increase GluR1 phosphorylation, and further prevent Aß-induced AMPAR endocytosis.166 However, some believed Aß-induced caspase-3 activity enhancement would cause dephosphorylation of GluR1 via calcineurin and thus resulted in AMPAR degradation.167 The Aß effect on GluR1 subunit may be an explanation of why GluR1 knockdown, but not GluR2 knockdown, could prevent Aß toxicity on AMPAR-mediated EPSC enhancement.168 Beyond GluR1 subunit, Aß has also shown effect on GluR2 and GluR3 subunits. Human AD samples had fewer GluR2/3 subunits on neuronal membrane.169 What’s more, the phosphorylation of GluR2170 and GluR3 by Aß is essential for its role in causing synaptic impairment and memory dysfunction.171 CaMKII may also play an important role in Aß-AMPAR interaction, since CaMKII expression enhancement could rescue Aß-induced AMPAR deficit on its ionic current and response.165 Similar to NMDAR, research has shown Aß oligomer could induce overactivation of AMPAR, leading to a substantial calcium influx and subsequent neuronal oxidative stress.150 Furthermore, intracellular Aß oligomers have been shown to induce neuronal hyperexcitability through AMPAR-mediated current.172 In addition to Aß changing AMPAR downstream pathway, AMPAR also mediates Aß metabolism. Research has shown steady-state AMPAR activity could increase ISF Aß level.173 However, evoked AMPAR activity could reduce Aß level in a dose-dependent manner via a pathway including NMDAR and IL-6.173 Although treatments based on the manipulation of AMPAR are relatively limited, previous work has shown that hippocampal neurons lacking the GluR3 subunit were protected from Aβ-induced synaptic depression, spine loss, and impairment of LTP.171 The behavioral outcome matched this finding, as GluR3-deficient APP mice maintained normal memory despite Aβ overproduction.171 The inhibition of AMPAR desensitization, which is the ability of AMPARs change their conformation under prolonged exposure to glutamate to prevent neuronal overexcitation, by cyclothiazide rescued synaptic plasticity in AD model mice.162 The research on behavior level is lacking regarding AMPAR regulation.

6.3. mGluR5 receptors

mGluR5 is a subtype of metabotropic glutamate receptors that plays a crucial role in modulating neuronal excitability and synaptic plasticity across various brain regions.174 Cellular prion protein (PrPC) plays a center role in the interaction between Aß and mGluR5. Experiments have shown Aß can interact with mGluR5 through a connection mediated by the PrPC.175, 176 The Aβ-PrPC-mGluR5 complex not only activated Fyn kinase which led to dendritic spine loss,175 but also mediated synaptic plasticity alterations. Its effect on synaptic plasticity particularly suppressed LTP and facilitated LTD.139, 176 By preventing Aß binding to PrPC, the Aß-induced LTD facilitation could be blocked.176 On the other hand, research has shown mGluR5 binds to PrPC via intracellular protein mediators, including Homer1b/c, CaMPKII and tyrosine kinase 2ß. Aß oligomers have been reported to increase the phosphorylation level of these intracellular protein mediators and thus induce deficit of synaptic plasticity.177 Furthermore, Aß showed high binding affinity to mGluR5 via PrPC only in male mice and human brain samples, but not in female samples.178 This sex-specific characteristics of Aß-PrPC-mGluR5 complex may be an explanation of sex difference in AD. In addition to its interaction with PrPC, Aβ oligomer itself can also change the configuration of mGluR5 by promoting their clustering, which will lead to higher intracellular calcium concentration and further synaptic impairments.179

The manipulation of mGluR5 has shown promising results in AD treatment. 2-Methyl-6-(phenylethynyl)pyridine (MPEP), a commonly used mGLuR5 antagonist, has shown to prevent Aß oligomer-induced impairment in LTP induction;161, 180 while MTEP, another mGluR5 antagonist, rescued spine loss and memory dysfunction in AD model mice.175 In addition, chronic treatment of 2-Chloro-4-((2,5-dimethyl-1-(4-(trifluoromethoxy)phenyl)-1H-imidazol-4-yl)ethynyl)pyridine (CTEP), a long-lasting metabotropic mGluR5 inhibitor, reduced Aß level and rescued cognition dysfunction of AD model mice.181 However, this effect may be sex-specific, as a recent study suggested that the effect of CTEP only works on male mice.178 Enhancing mGluR5 activity by CDPPB, a mGluR5 positive allosteric modulator, can prevent Aß-induced neuronal loss, but unfortunately had little effect on rescuing memory deficit on 14-month-old AD model mice.182

VII. Interaction between GABA receptors and Aß pathology

Gamma-aminobutyric acid (GABA) receptors are the primary inhibitory neurotransmitter receptors in the central nervous system. There are two main types of GABA receptors: GABA-A receptors and GABA-B receptors. Each type plays a critical role in neural inhibition but operates through different mechanisms. GABA-A receptors are ionotropic receptors and ligand-gated ion channels. Their response is fast, allowing chlorine ions flowing into their central pore composed of 5 subunits from seven subunit subfamilies (i.e. α, β, …).183 This fast inhibitory neurotransmission makes them crucial for maintaining the balance between neuronal excitation and inhibition, influencing everything from encoding sensory signals to cognitive processing. GABA-B receptors, on the other hand, are G-protein coupled receptors consisting of B1 and B2 two subunits. Their response is slower, but more prolonged compared to GABA-A receptors.184

7.1. GABA-A receptors

In the context of Alzheimer’s disease, Aß is found to modify the subunit composition of GABA-A receptors. This modification included down-regulation of the α1 and γ2 subunits, along with an up-regulation of α2, β1, and γ1 subunits in the AD brain of mouse model and human patients.185, 186 This change in composition correlated with an increased EC50 of the GABA-A receptor for GABA in the AD brain,185 implicating a reduced receptor sensitivity. APP-PSEN1 mice exhibited neuronal hyperexcitability in the locus coeruleus, which may result from impaired function and reduced expression of the GABA-A receptor α3 subunit. These changes may be attributed to Aβ toxicity, as the GABA-A receptor α3 subunit has been shown to overlap with Aβ oligomer expression in both APP-PSEN1 mice and AD patients.187 Aß enhanced the inhibitory GABAergic tonic conductance and decreased the inhibitory postsynaptic current mediated by GABA-A receptors, leading to hippocampal dysfunction.186, 188 Further, Aβ oligomer disrupted the balance between glutamatergic and GABAergic systems, inducing neuronal hyperexcitation by increasing extracellular glutamate levels, likely through impaired uptake, which heightened glutamatergic activity and spontaneous EPSC frequency. Simultaneously, Aβ reduced the effectiveness of GABAergic inhibition, as shown by its effects being reversed by the GABA-A receptor antagonist picrotoxin.157

The interaction between Aß and GABAergic system has also been studied with GABA receptor modulators.189 As for the agonist, chronic stimulation of GABA-A receptors with muscimol showed neuroprotective effects against Aß-induced neurotoxicity, but this protection was lost when co-treated with the GABA-A receptor antagonist bicuculline.190, 191 Similarly, α5IA, an agonist for GABA-A receptors containing the α5 subunit, decreased Aß-induced cell loss and restored the expression change of different subunits in GABA-A receptor.192 In AD model mice having GABAergic system deficit, early treatment with gaboxadol, another GABA-A receptor agonist, could reverse hippocampal neuronal hyperexcitation and mildly restore cognitive function.186 In addition, Aß was shown to inhibit GABA-induced Cl current, suggesting another mechanism for Aß-induced neuronal hyperexcitation.193 These investigations clearly highlighted the therapeutic potential of GABA-A receptor agonists against Aß-induced impairment.

GABA-A receptor antagonists, such as pentylenetetrazole (PTZ), picrotoxin (PTX), and bicuculline, have been implicated in modulating Aβ-induced neuronal and cognitive deficits. Research showed that these antagonists can rescue Aβ-induced LTP deficits,157, 194 and restore the morphology and functionality impairment of adult-born granule cells by hAPP.195 In particular, prolonged PTX administration prevented memory deficits and mitigated the Aβ-induced upregulation of postsynaptic density protein 95 (PSD95), GluN2B and GABA-A α1 subunit in AD mouse models.194 However, these therapeutic implications should be interpreted cautiously, given the antagonists can also exacerbate Aβ-induced seizures196 and neutralize the neuroprotective effects of melatonin.197

7.2. GABA-B receptors

Unlike the GABA-A receptors, GABA-B receptors do not form ion channels but instead influence cells through secondary messengers. These receptors typically function as heterodimers, consisting of GABA-B1 and GABA-B2 subunits, where GABA-B1 binds with GABA and GABA-B2 couples the receptor to G proteins.198 Activation of GABA-B receptors leads to the inhibition of adenylate cyclase, decreased cAMP levels, and opening of potassium channels, which further contributes to neuronal hyperpolarization. Their modulation of synaptic transmission is vital for controlling neuronal excitability over longer periods compared to the fast-acting GABA-A receptors.

The density and composition of GABA-B receptor were found to be altered in AD mice and human samples. Both postsynaptic and presynaptic densities of the GABA-B receptor were reduced in the hippocampus of AD model mice,199 possibly contributing to the cognitive dysfunction associated with AD. On molecular level, a novel non-coding RNA termed 17A, upregulated in AD patients, increased Aβ synthesis and Aβ42/Aβ40 ratio200. 17A also promoted the expression of an alternative GABA-B receptor isoform by altering RNA polymerase splicing. Research showed that human APP can interact with presynaptic GABA-B receptor and lead to GABA release inhibition.201 Research showed that inhibiting GABA-B receptors could alleviate Aß-induced neuronal toxicity. For instance, the use of GABA-B receptor antagonists, such as CGP35348, has shown promising results in AD models. In a rat model, CGP35348 treatment alleviated memory impairment induced by acute Aß toxicity.202 Similarly, CGP36216, another antagonist acting on presynaptic GABA-B receptor, was able to mitigate the neuronal hyperexcitation induced by hAPP overexpression.201 However, researchers found no neuronal protective effect on Aß25–35-induced toxicity with the treatment of baclofen, a GABA-B receptor agonist.190

VII. Conclusion

Alzheimer’s disease, the leading cause of dementia, is a significant global health challenge with no definitive cure. It is characterized by the pathological accumulation of amyloid-β and tau, which lead to neural network imbalance, synaptic dysfunction and cognitive decline. Emerging research underscored the critical role of Aβ interactions with neural receptors in exacerbating these pathological processes. While some neuronal receptor-targeted treatments showed promise, the complexity of AD pathology demanded a comprehensive understanding of how these interactions influence disease progression.

This review aimed to highlight existing knowledge on the interactions between Aβ and neuronal receptors, focusing on the physiological, cognitive, and clinical consequences of receptor-targeted pharmacological interventions. By examining receptor-specific mechanisms—including adrenergic, acetylcholine, dopamine, 5-HT, and GABA receptors—this paper highlighted the potential of targeting receptor pathways to mitigate Aβ-induced pathology. Moreover, the review introduced various receptor manipulations with therapeutic potential, underscoring the necessity for continued research to optimize these strategies for clinical application. Future efforts could focus on understanding the complex dynamics of neurotransmitter receptor-Aβ interactions and developing neural device-based therapies for AD that modulate receptor activity without disrupting overall neural information processing.203

Table 1.

The effect of adrenergic receptor manipulation in AD

Receptor Agonist/Antagonist Treatment Biological Model Main Result Reference
ß2 AR Agonist Isoproterenol WT mice brain slices with oAß Reduced Aβ-induced hippocampal impairment 32
ß2 AR Agonist Clenbuterol APP/PS1 mice Increased α-secretase, reduced Aβ plaques, and promoted hippocampal neurogenesis, dendritic branching and memory 33, 34
ß2 AR Agonist Procaterol WT mice brain slices with oAß Enhanced LTP, and preventd Aß-induced synaptic dysfunction 35
ß2 AR Agonist - Human subjects Reduced the risk of developing AD 36
ß2 AR Agonist Isoproterenol and clenbuterol APP/PS1 mice Enhanced γ-secretase activity, and increased cerebral amyloid plaque 38
ß2 AR Antagonist ICI118551 APP/PS1 mice Decreased cerebral amyloid plaque 38
ß2 AR Antagonist ICI118551 3xTg-AD mice Increased amyloidogenic APP processing, and exacerbated cognitive impairment 30
ß2 AR Antagonist ICI118551 AD-TG mice Inhibited dendritic ramification, downregulated α-secretase activity, and exacerbated cognitive deficit 39
α2 AR Agonist Clonidine APP/PS1 mice Exacerbated Aß production by promoting APP dislocation and cleavage 42
α2 AR Antagonist Idazoxan APP/PS1 mice Reduced Aß42, and mitigated memory impairment 42

Table 2.

The effect of cholinergic receptor manipulation in AD

Receptor Agonist/Antagonist Treatment Biologcal Model Main Result Reference
α7 nAchR Agonist Nicotine, epibatidine SK-N-MC cell culture with Aß42 Protected α7nAchR loss from Aβ42 54
α7 nAchR Agonist A-582941 3xTg-AD mice Increased c-Fos & BDNF expression, and improved cognition 69
α7 nAchR Antagonist methyllycaconitine HEK293T & SH-SY5Y cell culture with Aß42 Prevented Aß binding to α7nAchR, and prevented LDH release 55, 61
α7 nAchR Antagonist α-Bungarotoxin SH-SY5Y cell culture with Aß42 Diminished Aß42-associated mitochondrial dysfunction 71
M1 mAchR Agonist AF267B 3xTg-AD mice Reduced amyloid and Tau production, and improved memory performance 86
M1 mAchR Antagonist dicyclomine 3xTg-AD mice Shifted APP processing to amyloidogenic pathway (Higher BACE1 activity) 86
mAchR Agonist Oxo-M Rat frontal cortical brain slices with Aß25–35 Eliminated Aß effects on PKC & CaMKII 87
mAchR Antagonist Atropine; pirenzepine Wistar rat medial septal brain slices with Aß40, Aß25–35 Blocked Aß toxicity on glutamatergic synaptic transmission 51

Table 3.

The effect of dopaminergic receptor manipulation in AD

Receptor Agonist/Antagonist Treatment Biologcal Model Main Result Reference
D1R Agonist SKF38393 Icv Aß42-injected ICR mice Enhanced BDNF and Bcl-2 expression, reduced BACE1 activity and mitigated cognitive deficits 96
D1R Agonist A-68930 Icv Aß42-injected ICR mice Alleviated neuroinflammation, and mitigated cognitive deficits 95
D1R Antagonist SCH23390 Icv Aß42-injected ICR mice Exacerbated cognitive deficits 96
D2R Agonist Bromocriptine Icv Aß42-injected ICR mice Improved cognitive performance 97
D2/D3R Agonist Rotigotine AD patients Increased cortical excitability and restored central cholinergic transmission 98

Table 4.

The effect of 5-HT receptor manipulation in AD

Receptor Agonist/Antagonist Treatment Biological Model Main Result Reference
5-HT1AR Antagonist NAD-299 STZ-induced Wistar rats Reduced oxidative stress, decreased neuronal loss, and mitigated memory deficits 108, 109
5-HT1AR Antagonist WAY100635 Aβ42-injected WT mice Alleviated Aβ-induced learning and memory decline 110
5-HT2AR Agonist TCB-2 STZ-induced Wistar rats Reduced oxidative stress, decreased neuronal loss, and mitigated memory deficits 108, 109
5-HT2AR Antagonist Desloratadine APP/PS1 mice Reduced Amyloid plaque deposition by enhancing microglial Aß phagocytosis 116
5-HT2BR Antagonist MW701 Aβ42-injected WT mice Mitigated Aß42-induced impairments in LTP and memory deficits 117
5-HT4AR Agonist RS-67333 5xFAD mice Reduced Aß load and neuroinflammation, improved memory performance, 119, 121
5-HT6AR Antagonist SB-258585 STZ-induced Wistar rats, Aβ42-injected Wistar rats Attenuated LTP impairment and memory loss 126, 128
5-HT6AR Agonist WAY-181187 MK-801-induced Wistar rats Enhanced BDNF expression and prevented memory impairments 130
5-HT7R Agonist AS19 Aß-injected Wistar rats, STZ-induced Wistar rats Reduced Aß plaque deposition and neuronal apoptosis, improved memory performance 131, 132

Table 5.

The effect of glutamatergic receptor manipulation in AD

Receptor Agonist/Antagonist Treatment Biological Model Main Result Reference
NMDAR Agonist NMDA Cortical neuron culture of WT mice Increased α-secretase activity and inhibited amyloid generation 146
NMDAR Agonist NMDA Cortical neuron culture of WT mice Increased Aß production via extrasynaptic NMDAR activation to shift APP isoform to KPI-APP 147
NMDAR Antagonist Memantine AD patients Inhibited Aß generation and prevented memory dysfunction 151, 152
NMDAR Antagonist AP5 Hippocampal WT brain slices with Aß40, cortical neuron culture of WT mice Prevented the Aβo-mediated impairment of LTP and calcium imbalance 157, 158
NMDAR Antagonist CPP APP/PS1 mice, SD rat brain slices with oAß Increased ICF Aß level, and prevented spine loss 143, 159
NMDAR GluN2B Antagonist Ifenprodil, Ro 25–6981 Hippocampal neuron culture and slices of WT rats and mice Prevented Aß-induced LTP inhibition 149, 160, 161
mGluR5 Agonist CDPPB T41 mice Prevented Aß-induced neuronal loss, and reduced gliosis 182
mGluR5 Antagonist MPEP Hippocampal brain slices of WT mice and rat with oAß Prevented Aß-induced LTP impairment 161, 180
mGluR5 Antagonist MTEP APP/PS1 mice Rescued spine loss and memory dysfunction 175
mGluR5 Antagonist CTEP APP/PS1 mice Reduced Aß level and rescued cognition dysfunction 181

Table 6.

The effect of GABAergic receptor manipulation in AD

Receptor Agonist/Antagonist Treatment Biological Model Main Result Reference
GABA-AR Agonist Muscimol SD rat cortical and hippocampal neuron culture Inhibited ROS generation and Aβ-induced neuronal apoptosis 190, 191
GABA-AR Agonist α5IA Hippocampal primary cell culture of WT mice Reduced Aβ-induced cell death 192
GABA-AR Agonist Gaboxadol 5XFAD mice Reversed hippocampal hyperexcitation and mildly restored cognitive function 186
GABA-AR Antagonist Bicuculline, picrotoxin APP/PS1 mice Increased LTP level, and rescued memory deficit 194
GABA-AR Antagonist Picrotoxin WT mouse hippocampal brain slices with oAß Reduced soluble Aβo induced LTP deficit 157
GABA-AR Antagonist Picrotoxin hAPP transgenic mice Normalized the development of adult-born granule cells in hAPP mice 195
GABA-BR Antagonist CGP36216 hAPP transgenic mice Rescued neuronal hyperexcitation 201
GABA-BR Antagonist CGP35348 Icv Aß-injected Wistar rats Alleviated memory impairment 202

Acknowledgments

The authors have no acknowledgments to report.

Funding statement

This work was supported by NIH R01AG075114, R01NS119813, and U01AG066722.

Footnotes

Ethical considerations

This study was based on previously published data and did not involve any new experiments requiring ethical approval.

Consent to participate

This study did not involve any human subjects.

Consent for publication

All authors have given their consent for publication.

Declaration of conflicting interests

Q.W. is the co-founder of Sharper Sense, a company developing methods of enhancing sensory processing with neural interfaces.

Data availability

This study did not involve the collection or generation of new data.

References

  • 1.Zhao J, Liu X, Xia W, et al. Targeting Amyloidogenic Processing of APP in Alzheimer’s Disease. Front Mol Neurosci 2020; 13: 137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Anderson JP, Esch FS, Keim PS, et al. Exact cleavage site of Alzheimer amyloid precursor in neuronal PC-12 cells. Neuroscience letters 1991; 128: 126–128. [DOI] [PubMed] [Google Scholar]
  • 3.Wilson CA, Doms RW and Lee VM. Intracellular APP processing and Aβ production in Alzheimer disease. Journal of neuropathology and experimental neurology 1999; 58: 787–794. [DOI] [PubMed] [Google Scholar]
  • 4.Burdick D, Soreghan B, Kwon M, et al. Assembly and aggregation properties of synthetic Alzheimer’s A4/beta amyloid peptide analogs. Journal of Biological Chemistry 1992; 267: 546–554. [PubMed] [Google Scholar]
  • 5.Sherrington R, Rogaev E, Liang Ya, et al. Cloning of a gene bearing missense mutations in early-onset familial Alzheimer’s disease. Nature 1995; 375: 754–760. [DOI] [PubMed] [Google Scholar]
  • 6.De Jonghe C, Esselens C, Kumar-Singh S, et al. Pathogenic APP mutations near the γ-secretase cleavage site differentially affect Aβ secretion and APP C-terminal fragment stability. Human molecular genetics 2001; 10: 1665–1671. [DOI] [PubMed] [Google Scholar]
  • 7.Chen W, Gamache E, Rosenman DJ, et al. Familial Alzheimer’s mutations within APPTM increase Aβ42 production by enhancing accessibility of ε-cleavage site. Nature communications 2014; 5: 3037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Lambert MP, Barlow A, Chromy BA, et al. Diffusible, nonfibrillar ligands derived from Aβ1–42 are potent central nervous system neurotoxins. Proceedings of the National Academy of Sciences 1998; 95: 6448–6453. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Kayed R, Head E, Thompson JL, et al. Common structure of soluble amyloid oligomers implies common mechanism of pathogenesis. Science 2003; 300: 486–489. [DOI] [PubMed] [Google Scholar]
  • 10.Walsh DM, Klyubin I, Fadeeva JV, et al. Naturally secreted oligomers of amyloid β protein potently inhibit hippocampal long-term potentiation in vivo. Nature 2002; 416: 535–539. [DOI] [PubMed] [Google Scholar]
  • 11.Wilcox KC, Lacor PN, Pitt J, et al. Aβ oligomer-induced synapse degeneration in Alzheimer’s disease. Cellular and molecular neurobiology 2011; 31: 939–948. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Tomiyama T, Matsuyama S, Iso H, et al. A mouse model of amyloid β oligomers: their contribution to synaptic alteration, abnormal tau phosphorylation, glial activation, and neuronal loss in vivo. Journal of Neuroscience 2010; 30: 4845–4856. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Weiss E, Kann M and Wang Q. Neuromodulation of Neural Oscillations in Health and Disease. Biology 2023; 12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Sabatini BL and Tian L. Imaging Neurotransmitter and Neuromodulator Dynamics In Vivo with Genetically Encoded Indicators. Neuron 2020; 108: 17–32. [DOI] [PubMed] [Google Scholar]
  • 15.McCormick DA, Wang Z and Huguenard J. Neurotransmitter control of neocortical neuronal activity and excitability. Cereb Cortex 1993; 3: 387–398. [DOI] [PubMed] [Google Scholar]
  • 16.Delis I, Dmochowski JP, Sajda P, et al. Correlation of neural activity with behavioral kinematics reveals distinct sensory encoding and evidence accumulation processes during active tactile sensing. NeuroImage 2018; 175: 12–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Zheng HJV, Wang Q and Stanley GB. Adaptive shaping of cortical response selectivity in the vibrissa pathway. Journal of Neurophysiology 2015; 113: 3850–3865. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Sharma K, Pradhan S, Duffy LK, et al. Role of Receptors in Relation to Plaques and Tangles in Alzheimer’s Disease Pathology. Int J Mol Sci 2021; 22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Zhao J, Deng Y, Jiang Z, et al. G protein-coupled receptors (GPCRs) in Alzheimer’s disease: a focus on BACE1 related GPCRs. Frontiers in aging neuroscience 2016; 8: 58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Clewett D, Huang R, Velasco R, et al. Locus coeruleus activity strengthens prioritized memories under arousal. J Neurosci 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Totah NKB, Logothetis NK and Eschenko O. Noradrenergic ensemble-based modulation of cognition over multiple timescales. Brain Research 2019; 1709: 50–66. [DOI] [PubMed] [Google Scholar]
  • 22.Rodenkirch C, Liu Y, Schriver BJ, et al. Locus coeruleus activation enhances thalamic feature selectivity via norepinephrine regulation of intrathalamic circuit dynamics. Nature Neuroscience 2019; 22: 120–133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Structure Strosberg A., function, and regulation of adrenergic receptors. Protein Science 1993; 2: 1198–1209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Slater C and Wang Q. Alzheimer’s disease: An evolving understanding of noradrenergic involvement and the promising future of electroceutical therapies. Clin Transl Med 2021; 11: e397. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Chalermpalanupap T, Schroeder JP, Rorabaugh JM, et al. Locus Coeruleus Ablation Exacerbates Cognitive Deficits, Neuropathology, and Lethality in P301S Tau Transgenic Mice. J Neurosci 2018; 38: 74–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Jacobs Heidi IL, Becker John A, Kwong K, et al. In vivo and neuropathology data support locus coeruleus integrity as indicator of Alzheimer’s disease pathology and cognitive decline. Science Translational Medicine 2021; 13: eabj2511. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Manaye KF, Mouton PR, Xu G, et al. Age-related loss of noradrenergic neurons in the brains of triple transgenic mice. Age (Dordr) 2013; 35: 139–147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Goodman AM, Langner BM, Jackson N, et al. Heightened Hippocampal beta-Adrenergic Receptor Function Drives Synaptic Potentiation and Supports Learning and Memory in the TgF344-AD Rat Model during Prodromal Alzheimer’s Disease. J Neurosci 2021; 41: 5747–5761. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Tamano H, Ishikawa Y, Shioya A, et al. Adrenergic beta receptor activation reduces amyloid beta(1–42)-mediated intracellular Zn(2+) toxicity in dentate granule cells followed by rescuing impairment of dentate gyrus LTP. Neurotoxicology 2020; 79: 177–183. [DOI] [PubMed] [Google Scholar]
  • 30.Branca C, Wisely EV, Hartman LK, et al. Administration of a selective beta2 adrenergic receptor antagonist exacerbates neuropathology and cognitive deficits in a mouse model of Alzheimer’s disease. Neurobiol Aging 2014; 35: 2726–2735. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Wang D, Govindaiah G, Liu R, et al. Binding of amyloid beta peptide to beta2 adrenergic receptor induces PKA-dependent AMPA receptor hyperactivity. FASEB J 2010; 24: 3511–3521. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Li S, Jin M, Zhang D, et al. Environmental novelty activates beta2-adrenergic signaling to prevent the impairment of hippocampal LTP by Abeta oligomers. Neuron 2013; 77: 929–941. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Chai GS, Wang YY, Yasheng A, et al. Beta 2-adrenergic receptor activation enhances neurogenesis in Alzheimer’s disease mice. Neural Regen Res 2016; 11: 1617–1624. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Chai GS, Wang YY, Zhu D, et al. Activation of beta(2)-adrenergic receptor promotes dendrite ramification and spine generation in APP/PS1 mice. Neurosci Lett 2017; 636: 158–164. [DOI] [PubMed] [Google Scholar]
  • 35.Jin M, Wei Z, Ramalingam N, et al. Activation of beta(2)-adrenergic receptors prevents AD-type synaptotoxicity via epigenetic mechanisms. Mol Psychiatry 2023; 28: 4877–4888. [DOI] [PubMed] [Google Scholar]
  • 36.Hutten DR, Bos JHJ, de Vos S, et al. Targeting the Beta-2-Adrenergic Receptor and the Risk of Developing Alzheimer’s Disease: A Retrospective Inception Cohort Study. J Alzheimers Dis 2022; 87: 1089–1101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Yu NN, Wang XX, Yu JT, et al. Blocking beta2-adrenergic receptor attenuates acute stress-induced amyloid beta peptides production. Brain Res 2010; 1317: 305–310. [DOI] [PubMed] [Google Scholar]
  • 38.Ni Y, Zhao X, Bao G, et al. Activation of beta2-adrenergic receptor stimulates gamma-secretase activity and accelerates amyloid plaque formation. Nat Med 2006; 12: 1390–1396. [DOI] [PubMed] [Google Scholar]
  • 39.Wu Q, Sun JX, Song XH, et al. Blocking beta 2-adrenergic receptor inhibits dendrite ramification in a mouse model of Alzheimer’s disease. Neural Regen Res 2017; 12: 1499–1506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Zhang F, Gannon M, Chen Y, et al. beta-amyloid redirects norepinephrine signaling to activate the pathogenic GSK3beta/tau cascade. Sci Transl Med 2020; 12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Zhang F, Gannon M, Chen Y, et al. The amyloid precursor protein modulates alpha(2A)-adrenergic receptor endocytosis and signaling through disrupting arrestin 3 recruitment. FASEB J 2017; 31: 4434–4446. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Chen Y, Peng Y, Che P, et al. alpha(2A) adrenergic receptor promotes amyloidogenesis through disrupting APP-SorLA interaction. Proc Natl Acad Sci U S A 2014; 111: 17296–17301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Infantino R, Boccella S, Scuteri D, et al. 2-pentadecyl-2-oxazoline prevents cognitive and social behaviour impairments in the Amyloid beta-induced Alzheimer-like mice model: Bring the alpha2 adrenergic receptor back into play. Biomed Pharmacother 2022; 156: 113844. [DOI] [PubMed] [Google Scholar]
  • 44.Gibbs ME, Maksel D, Gibbs Z, et al. Memory loss caused by beta-amyloid protein is rescued by a beta(3)-adrenoceptor agonist. Neurobiol Aging 2010; 31: 614–624. [DOI] [PubMed] [Google Scholar]
  • 45.Tournissac M, Vu TM, Vrabic N, et al. Repurposing beta-3 adrenergic receptor agonists for Alzheimer’s disease: beneficial effects in a mouse model. Alzheimers Res Ther 2021; 13: 103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Yu ZY, Yi X, Wang YR, et al. Inhibiting alpha1-adrenergic receptor signaling pathway ameliorates AD-type pathologies and behavioral deficits in APPswe/PS1 mouse model. J Neurochem 2022; 161: 293–307. [DOI] [PubMed] [Google Scholar]
  • 47.Slater C, Liu Y, Weiss E, et al. The Neuromodulatory Role of the Noradrenergic and Cholinergic Systems and Their Interplay in Cognitive Functions: A Focused Review. Brain Sciences 2022; 12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Jiang S, Li Y, Zhang C, et al. M1 muscarinic acetylcholine receptor in Alzheimer’s disease. Neurosci Bull 2014; 30: 295–307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Wu J and Lukas RJ. Naturally-expressed nicotinic acetylcholine receptor subtypes. Biochemical pharmacology 2011; 82: 800–807. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Chen GJ, Xiong Z and Yan Z. Abeta impairs nicotinic regulation of inhibitory synaptic transmission and interneuron excitability in prefrontal cortex. Mol Neurodegener 2013; 8: 3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Santos-Torres J, Fuente A, Criado JM, et al. Glutamatergic synaptic depression by synthetic amyloid beta-peptide in the medial septum. J Neurosci Res 2007; 85: 634–648. [DOI] [PubMed] [Google Scholar]
  • 52.Dougherty JJ, Wu J and Nichols RA. Beta-amyloid regulation of presynaptic nicotinic receptors in rat hippocampus and neocortex. J Neurosci 2003; 23: 6740–6747. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Borroni V and Barrantes FJ. Homomeric and heteromeric α7 nicotinic acetylcholine receptors in health and some central nervous system diseases. Membranes 2021; 11: 664. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Wang HY, Lee DH, D’Andrea MR, et al. beta-Amyloid(1–42) binds to alpha7 nicotinic acetylcholine receptor with high affinity. Implications for Alzheimer’s disease pathology. J Biol Chem 2000; 275: 5626–5632. [DOI] [PubMed] [Google Scholar]
  • 55.Cecon E, Dam J, Luka M, et al. Quantitative assessment of oligomeric amyloid beta peptide binding to alpha7 nicotinic receptor. Br J Pharmacol 2019; 176: 3475–3488. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Lasala M, Fabiani C, Corradi J, et al. Molecular Modulation of Human alpha7 Nicotinic Receptor by Amyloid-beta Peptides. Front Cell Neurosci 2019; 13: 37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Spencer JP, Weil A, Hill K, et al. Transgenic mice over-expressing human beta-amyloid have functional nicotinic alpha 7 receptors. Neuroscience 2006; 137: 795–805. [DOI] [PubMed] [Google Scholar]
  • 58.Maatuk N and Samson AO. Modeling the binding mechanism of Alzheimer’s Abeta1-42 to nicotinic acetylcholine receptors based on similarity with snake alpha-neurotoxins. Neurotoxicology 2013; 34: 236–242. [DOI] [PubMed] [Google Scholar]
  • 59.Ashenafi S, Fuente A, Criado JM, et al. Beta-Amyloid peptide25–35 depresses excitatory synaptic transmission in the rat basolateral amygdala “in vitro”. Neurobiol Aging 2005; 26: 419–428. [DOI] [PubMed] [Google Scholar]
  • 60.Liu Q, Xie X, Lukas RJ, et al. A novel nicotinic mechanism underlies beta-amyloid-induced neuronal hyperexcitation. J Neurosci 2013; 33: 7253–7263. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Liu Q, Xie X, Emadi S, et al. A novel nicotinic mechanism underlies beta-amyloid-induced neurotoxicity. Neuropharmacology 2015; 97: 457–463. [DOI] [PubMed] [Google Scholar]
  • 62.Chen L, Yamada K, Nabeshima T, et al. alpha7 Nicotinic acetylcholine receptor as a target to rescue deficit in hippocampal LTP induction in beta-amyloid infused rats. Neuropharmacology 2006; 50: 254–268. [DOI] [PubMed] [Google Scholar]
  • 63.He YX, Wu MN, Zhang H, et al. Amyloid beta-protein suppressed nicotinic acetylcholine receptor-mediated currents in acutely isolated rat hippocampal CA1 pyramidal neurons. Synapse 2013; 67: 11–20. [DOI] [PubMed] [Google Scholar]
  • 64.Hahm ET, Nagaraja RY, Waro G, et al. Cholinergic Homeostatic Synaptic Plasticity Drives the Progression of Abeta-Induced Changes in Neural Activity. Cell Rep 2018; 24: 342–354. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Stoiljkovic M, Kelley C, Hajos GP, et al. Hippocampal network dynamics in response to alpha7 nACh receptors activation in amyloid-beta overproducing transgenic mice. Neurobiol Aging 2016; 45: 161–168. [DOI] [PubMed] [Google Scholar]
  • 66.Ju Y, Asahi T and Sawamura N. Arctic mutant Abeta40 aggregates on alpha7 nicotinic acetylcholine receptors and inhibits their functions. J Neurochem 2014; 131: 667–674. [DOI] [PubMed] [Google Scholar]
  • 67.Fonar G, Polis B, Sams DS, et al. Modified Snake alpha-Neurotoxin Averts beta-Amyloid Binding to alpha7 Nicotinic Acetylcholine Receptor and Reverses Cognitive Deficits in Alzheimer’s Disease Mice. Mol Neurobiol 2021; 58: 2322–2341. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Soderman A, Mikkelsen JD, West MJ, et al. Activation of nicotinic alpha(7) acetylcholine receptor enhances long term potentation in wild type mice but not in APP(swe)/PS1DeltaE9 mice. Neurosci Lett 2011; 487: 325–329. [DOI] [PubMed] [Google Scholar]
  • 69.Medeiros R, Castello NA, Cheng D, et al. alpha7 Nicotinic receptor agonist enhances cognition in aged 3xTg-AD mice with robust plaques and tangles. Am J Pathol 2014; 184: 520–529. [DOI] [PubMed] [Google Scholar]
  • 70.Ni R, Marutle A and Nordberg A. Modulation of alpha7 nicotinic acetylcholine receptor and fibrillar amyloid-beta interactions in Alzheimer’s disease brain. J Alzheimers Dis 2013; 33: 841–851. [DOI] [PubMed] [Google Scholar]
  • 71.Sinclair P and Kabbani N. Nicotinic receptor components of amyloid beta 42 proteome regulation in human neural cells. PLoS One 2022; 17: e0270479. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Tropea MR, Li Puma DD, Melone M, et al. Genetic deletion of alpha7 nicotinic acetylcholine receptors induces an age-dependent Alzheimer’s disease-like pathology. Prog Neurobiol 2021; 206: 102154. [DOI] [PubMed] [Google Scholar]
  • 73.Dziewczapolski G, Glogowski CM, Masliah E, et al. Deletion of the alpha 7 nicotinic acetylcholine receptor gene improves cognitive deficits and synaptic pathology in a mouse model of Alzheimer’s disease. J Neurosci 2009; 29: 8805–8815. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Moretti M, Zoli M, George AA, et al. The novel α7β2-nicotinic acetylcholine receptor subtype is expressed in mouse and human basal forebrain: biochemical and pharmacological characterization. Molecular pharmacology 2014; 86: 306–317. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Wu J, Liu Q, Tang P, et al. Heteromeric alpha7beta2 Nicotinic Acetylcholine Receptors in the Brain. Trends Pharmacol Sci 2016; 37: 562–574. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Liu Q, Huang Y, Xue F, et al. A novel nicotinic acetylcholine receptor subtype in basal forebrain cholinergic neurons with high sensitivity to amyloid peptides. J Neurosci 2009; 29: 918–929. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Liu Q, Huang Y, Shen J, et al. Functional alpha7beta2 nicotinic acetylcholine receptors expressed in hippocampal interneurons exhibit high sensitivity to pathological level of amyloid beta peptides. BMC Neurosci 2012; 13: 155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.George AA, Vieira JM, Xavier-Jackson C, et al. Implications of Oligomeric Amyloid-Beta (oAbeta(42)) Signaling through alpha7beta2-Nicotinic Acetylcholine Receptors (nAChRs) on Basal Forebrain Cholinergic Neuronal Intrinsic Excitability and Cognitive Decline. J Neurosci 2021; 41: 555–575. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Grupe M, Grunnet M, Bastlund JF, et al. Targeting α4β2 nicotinic acetylcholine receptors in central nervous system disorders: perspectives on positive allosteric modulation as a therapeutic approach. Basic & clinical pharmacology & toxicology 2015; 116: 187–200. [DOI] [PubMed] [Google Scholar]
  • 80.Dineley KT, Pandya AA and Yakel JL. Nicotinic ACh receptors as therapeutic targets in CNS disorders. Trends in pharmacological sciences 2015; 36: 96–108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Wu J, Kuo YP, George AA, et al. beta-Amyloid directly inhibits human alpha4beta2-nicotinic acetylcholine receptors heterologously expressed in human SH-EP1 cells. J Biol Chem 2004; 279: 37842–37851. [DOI] [PubMed] [Google Scholar]
  • 82.Salehi B, Sestito S, Rapposelli S, et al. Epibatidine: a promising natural alkaloid in health. Biomolecules 2018; 9: 6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Olivero G, Grilli M, Chen J, et al. Effects of soluble beta-amyloid on the release of neurotransmitters from rat brain synaptosomes. Front Aging Neurosci 2014; 6: 166. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Roberts JP, Stokoe SA, Sathler MF, et al. Selective coactivation of alpha7- and alpha4beta2-nicotinic acetylcholine receptors reverses beta-amyloid-induced synaptic dysfunction. J Biol Chem 2021; 296: 100402. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Felder CC, Bymaster FP, Ward J, et al. Therapeutic opportunities for muscarinic receptors in the central nervous system. Journal of medicinal chemistry 2000; 43: 4333–4353. [DOI] [PubMed] [Google Scholar]
  • 86.Caccamo A, Oddo S, Billings LM, et al. M1 receptors play a central role in modulating AD-like pathology in transgenic mice. Neuron 2006; 49: 671–682. [DOI] [PubMed] [Google Scholar]
  • 87.Gu Z, Zhong P and Yan Z. Activation of muscarinic receptors inhibits beta-amyloid peptide-induced signaling in cortical slices. J Biol Chem 2003; 278: 17546–17556. [DOI] [PubMed] [Google Scholar]
  • 88.Janickova H, Rudajev V, Zimcik P, et al. Uncoupling of M1 muscarinic receptor/G-protein interaction by amyloid beta(1–42). Neuropharmacology 2013; 67: 272–283. [DOI] [PubMed] [Google Scholar]
  • 89.Missale C, Nash SR, Robinson SW, et al. Dopamine receptors: from structure to function. Physiological reviews 1998. [DOI] [PubMed] [Google Scholar]
  • 90.Hisahara S and Shimohama S. Dopamine Receptors and Parkinson′ s Disease. International journal of medicinal chemistry 2011; 2011: 403039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Seeman P and Niznik HB. Dopamine receptors and transporters in Parkinson’s disease and schizophrenia. The FASEB Journal 1990; 4: 2737–2744. [DOI] [PubMed] [Google Scholar]
  • 92.Moreno-Castilla P, Rodriguez-Duran LF, Guzman-Ramos K, et al. Dopaminergic neurotransmission dysfunction induced by amyloid-β transforms cortical long-term potentiation into long-term depression and produces memory impairment. Neurobiology of aging 2016; 41: 187–199. [DOI] [PubMed] [Google Scholar]
  • 93.Kumar U and Patel SC. Immunohistochemical localization of dopamine receptor subtypes (D1R–D5R) in Alzheimer’s disease brain. Brain research 2007; 1131: 187–196. [DOI] [PubMed] [Google Scholar]
  • 94.Kemppainen N, Laine M, Laakso M, et al. Hippocampal dopamine D2 receptors correlate with memory functions in Alzheimer’s disease. European journal of neuroscience 2003; 18: 149–154. [DOI] [PubMed] [Google Scholar]
  • 95.Cheng Z-Y, Xia Q-P, Hu Y-H, et al. Dopamine D1 receptor agonist A-68930 ameliorates Aβ1–42-induced cognitive impairment and neuroinflammation in mice. International Immunopharmacology 2020; 88: 106963. [DOI] [PubMed] [Google Scholar]
  • 96.Zang X, Cheng Z-Y, Sun Y, et al. The ameliorative effects and underlying mechanisms of dopamine D1-like receptor agonist SKF38393 on Aβ1–42-induced cognitive impairment. Progress in Neuro-Psychopharmacology and Biological Psychiatry 2018; 81: 250–261. [DOI] [PubMed] [Google Scholar]
  • 97.Liu X, Cheng Z-Y, Li Y-F, et al. Dopamine D2 receptor agonist Bromocriptine ameliorates Aβ1–42-induced memory deficits and neuroinflammation in mice. European Journal of Pharmacology 2023; 938: 175443. [DOI] [PubMed] [Google Scholar]
  • 98.Martorana A, Di Lorenzo F, Esposito Z, et al. Dopamine D2-agonist Rotigotine effects on cortical excitability and central cholinergic transmission in Alzheimer’s disease patients. Neuropharmacology 2013; 64: 108–113. [DOI] [PubMed] [Google Scholar]
  • 99.Sharp T and Barnes NM. Central 5-HT receptors and their function; present and future. Neuropharmacology 2020; 177: 108155. [DOI] [PubMed] [Google Scholar]
  • 100.Hannon J and Hoyer D. Molecular biology of 5-HT receptors. Behavioural brain research 2008; 195: 198–213. [DOI] [PubMed] [Google Scholar]
  • 101.Carhart-Harris RL and Nutt DJ. Serotonin and brain function: a tale of two receptors. Journal of psychopharmacology 2017; 31: 1091–1120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Eremin DV, Kondaurova EM, Rodnyy AY, et al. Serotonin Receptors as a Potential Target in the Treatment of Alzheimer’s Disease. Biochemistry (Mosc) 2023; 88: 2023–2042. [DOI] [PubMed] [Google Scholar]
  • 103.Vidal B, Sebti J, Verdurand M, et al. Agonist and antagonist bind differently to 5-HT1A receptors during Alzheimer’s disease: A post-mortem study with PET radiopharmaceuticals. Neuropharmacology 2016; 109: 88–95. [DOI] [PubMed] [Google Scholar]
  • 104.Mattsson P, Cselenyi Z, Andree B, et al. Decreased 5-HT(1A) binding in mild Alzheimer’s disease-A positron emission tomography study. Synapse 2022; 76: e22235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Verdurand M, Berod A, Le Bars D, et al. Effects of amyloid-beta peptides on the serotoninergic 5-HT1A receptors in the rat hippocampus. Neurobiol Aging 2011; 32: 103–114. [DOI] [PubMed] [Google Scholar]
  • 106.Truchot L, Costes SN, Zimmer L, et al. Up-regulation of hippocampal serotonin metabolism in mild cognitive impairment. Neurology 2007; 69: 1012–1017. [DOI] [PubMed] [Google Scholar]
  • 107.Verdurand M, Chauveau F, Daoust A, et al. Differential effects of amyloid-beta 1–40 and 1–42 fibrils on 5-HT1A serotonin receptors in rat brain. Neurobiol Aging 2016; 40: 11–21. [DOI] [PubMed] [Google Scholar]
  • 108.Afshar S, Shahidi S, Rohani AH, et al. The effect of NAD-299 and TCB-2 on learning and memory, hippocampal BDNF levels and amyloid plaques in Streptozotocin-induced memory deficits in male rats. Psychopharmacology (Berl) 2018; 235: 2809–2822. [DOI] [PubMed] [Google Scholar]
  • 109.Afshar S, Shahidi S, Rohani AH, et al. Protective effects of 5-HT(1A) receptor antagonist and 5-HT(2A) receptor agonist on the biochemical and histological features in a rat model of Alzheimer’s disease. J Chem Neuroanat 2019; 96: 140–147. [DOI] [PubMed] [Google Scholar]
  • 110.Wang M, Zong HF, Chang KW, et al. 5-HT(1A)R alleviates Abeta-induced cognitive decline and neuroinflammation through crosstalk with NF-kappaB pathway in mice. Int Immunopharmacol 2020; 82: 106354. [DOI] [PubMed] [Google Scholar]
  • 111.Marner L, Frokjaer VG, Kalbitzer J, et al. Loss of serotonin 2A receptors exceeds loss of serotonergic projections in early Alzheimer’s disease: a combined [11C]DASB and [18F]altanserin-PET study. Neurobiol Aging 2012; 33: 479–487. [DOI] [PubMed] [Google Scholar]
  • 112.Hasselbalch SG, Madsen K, Svarer C, et al. Reduced 5-HT2A receptor binding in patients with mild cognitive impairment. Neurobiol Aging 2008; 29: 1830–1838. [DOI] [PubMed] [Google Scholar]
  • 113.Lai MK, Tsang SW, Alder JT, et al. Loss of serotonin 5-HT2A receptors in the postmortem temporal cortex correlates with rate of cognitive decline in Alzheimer’s disease. Psychopharmacology (Berl) 2005; 179: 673–677. [DOI] [PubMed] [Google Scholar]
  • 114.Christensen R, Marcussen AB, Wortwein G, et al. Abeta(1–42) injection causes memory impairment, lowered cortical and serum BDNF levels, and decreased hippocampal 5-HT(2A) levels. Exp Neurol 2008; 210: 164–171. [DOI] [PubMed] [Google Scholar]
  • 115.Holm P, Ettrup A, Klein AB, et al. Plaque deposition dependent decrease in 5-HT2A serotonin receptor in AbetaPPswe/PS1dE9 amyloid overexpressing mice. J Alzheimers Dis 2010; 20: 1201–1213. [DOI] [PubMed] [Google Scholar]
  • 116.Lu J, Zhang C, Lv J, et al. Antiallergic drug desloratadine as a selective antagonist of 5HT(2A) receptor ameliorates pathology of Alzheimer’s disease model mice by improving microglial dysfunction. Aging Cell 2021; 20: e13286. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Acquarone E, Argyrousi EK, Arancio O, et al. The 5HT2b Receptor in Alzheimer’s Disease: Increased Levels in Patient Brains and Antagonist Attenuation of Amyloid and Tau Induced Dysfunction. Journal of Alzheimer’s Disease 2024: 1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Anzalone M, Karam SA, Briting SR, et al. Serotonin-2B receptor (5-HT2BR) expression and binding in the brain of APPswe/PS1dE9 transgenic mice and in Alzheimer’s disease brain tissue. Neuroscience Letters 2025; 844: 138013. [DOI] [PubMed] [Google Scholar]
  • 119.Baranger K, Giannoni P, Girard SD, et al. Chronic treatments with a 5-HT(4) receptor agonist decrease amyloid pathology in the entorhinal cortex and learning and memory deficits in the 5xFAD mouse model of Alzheimer’s disease. Neuropharmacology 2017; 126: 128–141. [DOI] [PubMed] [Google Scholar]
  • 120.Quiedeville A, Boulouard M, Hamidouche K, et al. Chronic activation of 5-HT4 receptors or blockade of 5-HT6 receptors improve memory performances. Behav Brain Res 2015; 293: 10–17. [DOI] [PubMed] [Google Scholar]
  • 121.Giannoni P, Gaven F, de Bundel D, et al. Early administration of RS 67333, a specific 5-HT4 receptor agonist, prevents amyloidogenesis and behavioral deficits in the 5XFAD mouse model of Alzheimer’s disease. Front Aging Neurosci 2013; 5: 96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Nirogi R, Mohammed AR, Shinde AK, et al. Discovery and Preclinical Characterization of Usmarapride (SUVN-D4010): A Potent, Selective 5-HT(4) Receptor Partial Agonist for the Treatment of Cognitive Deficits Associated with Alzheimer’s Disease. J Med Chem 2021; 64: 10641–10665. [DOI] [PubMed] [Google Scholar]
  • 123.Fisher JR, Wallace CE, Tripoli DL, et al. Redundant Gs-coupled serotonin receptors regulate amyloid-beta metabolism in vivo. Mol Neurodegener 2016; 11: 45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Pimenova AA, Thathiah A, De Strooper B, et al. Regulation of amyloid precursor protein processing by serotonin signaling. PLoS One 2014; 9: e87014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Cochet M, Donneger R, Cassier E, et al. 5-HT4 receptors constitutively promote the non-amyloidogenic pathway of APP cleavage and interact with ADAM10. ACS Chem Neurosci 2013; 4: 130–140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Hashemi-Firouzi N, Shahidi S, Soleimani-Asl S, et al. 5-Hydroxytryptamine receptor 6 antagonist, SB258585 exerts neuroprotection in a rat model of Streptozotocin-induced Alzheimer’s disease. Metab Brain Dis 2018; 33: 1243–1253. [DOI] [PubMed] [Google Scholar]
  • 127.Ivachtchenko AV, Lavrovsky Y and Ivanenkov YA. AVN-211, Novel and Highly Selective 5-HT6 Receptor Small Molecule Antagonist, for the Treatment of Alzheimer’s Disease. Mol Pharm 2016; 13: 945–963. [DOI] [PubMed] [Google Scholar]
  • 128.Shahidi S, Hashemi-Firouzi N, Asl SS, et al. Serotonin type 6 receptor antagonist attenuates the impairment of long-term potentiation and memory induced by Abeta. Behav Brain Res 2019; 364: 205–212. [DOI] [PubMed] [Google Scholar]
  • 129.Hu L, Wang B and Zhang Y. Serotonin 5-HT6 receptors affect cognition in a mouse model of Alzheimer’s disease by regulating cilia function. Alzheimers Res Ther 2017; 9: 76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Rychtyk J, Partyka A, Gdula-Argasinska J, et al. 5-HT(6) receptor agonist and antagonist improve memory impairments and hippocampal BDNF signaling alterations induced by MK-801. Brain Res 2019; 1722: 146375. [DOI] [PubMed] [Google Scholar]
  • 131.Shahidi S, Asl SS, Komaki A, et al. The effect of chronic stimulation of serotonin receptor type 7 on recognition, passive avoidance memory, hippocampal long-term potentiation, and neuronal apoptosis in the amyloid beta protein treated rat. Psychopharmacology (Berl) 2018; 235: 1513–1525. [DOI] [PubMed] [Google Scholar]
  • 132.Hashemi-Firouzi N, Komaki A, Soleimani Asl S, et al. The effects of the 5-HT7 receptor on hippocampal long-term potentiation and apoptosis in a rat model of Alzheimer’s disease. Brain Res Bull 2017; 135: 85–91. [DOI] [PubMed] [Google Scholar]
  • 133.Schoepp DD. Novel functions for subtypes of metabotropic glutamate receptors. Neurochemistry international 1994; 24: 439–449. [DOI] [PubMed] [Google Scholar]
  • 134.Marcantoni A, Cerullo MS, Buxeda P, et al. Amyloid Beta42 oligomers up-regulate the excitatory synapses by potentiating presynaptic release while impairing postsynaptic NMDA receptors. J Physiol 2020; 598: 2183–2197. [DOI] [PubMed] [Google Scholar]
  • 135.Muller MK, Jacobi E, Sakimura K, et al. NMDA receptors mediate synaptic depression, but not spine loss in the dentate gyrus of adult amyloid Beta (Abeta) overexpressing mice. Acta Neuropathol Commun 2018; 6: 110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Taniguchi K, Yamamoto F, Amano A, et al. Amyloid-beta oligomers interact with NMDA receptors containing GluN2B subunits and metabotropic glutamate receptor 1 in primary cortical neurons: Relevance to the synapse pathology of Alzheimer’s disease. Neurosci Res 2022; 180: 90–98. [DOI] [PubMed] [Google Scholar]
  • 137.Olajide OJ and Chapman CA. Amyloid-beta (1–42) peptide induces rapid NMDA receptor-dependent alterations at glutamatergic synapses in the entorhinal cortex. Neurobiol Aging 2021; 105: 296–309. [DOI] [PubMed] [Google Scholar]
  • 138.Kessels HW, Nabavi S and Malinow R. Metabotropic NMDA receptor function is required for beta-amyloid-induced synaptic depression. Proc Natl Acad Sci U S A 2013; 110: 4033–4038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Shankar GM, Li S, Mehta TH, et al. Amyloid-beta protein dimers isolated directly from Alzheimer’s brains impair synaptic plasticity and memory. Nat Med 2008; 14: 837–842. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Raymond CR, Ireland DR and Abraham WC. NMDA receptor regulation by amyloid-beta does not account for its inhibition of LTP in rat hippocampus. Brain Res 2003; 968: 263–272. [DOI] [PubMed] [Google Scholar]
  • 141.Kervern M, Angeli A, Nicole O, et al. Selective impairment of some forms of synaptic plasticity by oligomeric amyloid-beta peptide in the mouse hippocampus: implication of extrasynaptic NMDA receptors. J Alzheimers Dis 2012; 32: 183–196. [DOI] [PubMed] [Google Scholar]
  • 142.Back MK, Ruggieri S, Jacobi E, et al. Amyloid Beta-Mediated Changes in Synaptic Function and Spine Number of Neocortical Neurons Depend on NMDA Receptors. Int J Mol Sci 2021; 22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Shankar GM, Bloodgood BL, Townsend M, et al. Natural oligomers of the Alzheimer amyloid-beta protein induce reversible synapse loss by modulating an NMDA-type glutamate receptor-dependent signaling pathway. J Neurosci 2007; 27: 2866–2875. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Tackenberg C, Grinschgl S, Trutzel A, et al. NMDA receptor subunit composition determines beta-amyloid-induced neurodegeneration and synaptic loss. Cell Death Dis 2013; 4: e608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Snyder EM, Nong Y, Almeida CG, et al. Regulation of NMDA receptor trafficking by amyloid-beta. Nat Neurosci 2005; 8: 1051–1058. [DOI] [PubMed] [Google Scholar]
  • 146.Hoey SE, Williams RJ and Perkinton MS. Synaptic NMDA receptor activation stimulates alpha-secretase amyloid precursor protein processing and inhibits amyloid-beta production. J Neurosci 2009; 29: 4442–4460. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Bordji K, Becerril-Ortega J, Nicole O, et al. Activation of extrasynaptic, but not synaptic, NMDA receptors modifies amyloid precursor protein expression pattern and increases amyloid-ss production. J Neurosci 2010; 30: 15927–15942. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Lesne S, Ali C, Gabriel C, et al. NMDA receptor activation inhibits alpha-secretase and promotes neuronal amyloid-beta production. J Neurosci 2005; 25: 9367–9377. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Sinnen BL, Bowen AB, Gibson ES, et al. Local and Use-Dependent Effects of beta-Amyloid Oligomers on NMDA Receptor Function Revealed by Optical Quantal Analysis. J Neurosci 2016; 36: 11532–11543. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Alberdi E, Sanchez-Gomez MV, Cavaliere F, et al. Amyloid beta oligomers induce Ca2+ dysregulation and neuronal death through activation of ionotropic glutamate receptors. Cell Calcium 2010; 47: 264–272. [DOI] [PubMed] [Google Scholar]
  • 151.Johnson JW and Kotermanski SE. Mechanism of action of memantine. Current opinion in pharmacology 2006; 6: 61–67. [DOI] [PubMed] [Google Scholar]
  • 152.Varadharajan A, Davis AD, Ghosh A, et al. Guidelines for pharmacotherapy in Alzheimer’s disease–A primer on FDA-approved drugs. Journal of neurosciences in rural practice 2023; 14: 566. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Xia P, Chen HS, Zhang D, et al. Memantine preferentially blocks extrasynaptic over synaptic NMDA receptor currents in hippocampal autapses. J Neurosci 2010; 30: 11246–11250. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Martinez-Coria H, Green KN, Billings LM, et al. Memantine improves cognition and reduces Alzheimer’s-like neuropathology in transgenic mice. Am J Pathol 2010; 176: 870–880. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Decker H, Lo KY, Unger SM, et al. Amyloid-beta peptide oligomers disrupt axonal transport through an NMDA receptor-dependent mechanism that is mediated by glycogen synthase kinase 3beta in primary cultured hippocampal neurons. J Neurosci 2010; 30: 9166–9171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Stazi M and Wirths O. Chronic Memantine Treatment Ameliorates Behavioral Deficits, Neuron Loss, and Impaired Neurogenesis in a Model of Alzheimer’s Disease. Mol Neurobiol 2021; 58: 204–216. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Lei M, Xu H, Li Z, et al. Soluble Abeta oligomers impair hippocampal LTP by disrupting glutamatergic/GABAergic balance. Neurobiol Dis 2016; 85: 111–121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Texido L, Martin-Satue M, Alberdi E, et al. Amyloid beta peptide oligomers directly activate NMDA receptors. Cell Calcium 2011; 49: 184–190. [DOI] [PubMed] [Google Scholar]
  • 159.Verges DK, Restivo JL, Goebel WD, et al. Opposing synaptic regulation of amyloid-beta metabolism by NMDA receptors in vivo. J Neurosci 2011; 31: 11328–11337. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Li S, Jin M, Koeglsperger T, et al. Soluble Abeta oligomers inhibit long-term potentiation through a mechanism involving excessive activation of extrasynaptic NR2B-containing NMDA receptors. J Neurosci 2011; 31: 6627–6638. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Rammes G, Hasenjager A, Sroka-Saidi K, et al. Therapeutic significance of NR2B-containing NMDA receptors and mGluR5 metabotropic glutamate receptors in mediating the synaptotoxic effects of beta-amyloid oligomers on long-term potentiation (LTP) in murine hippocampal slices. Neuropharmacology 2011; 60: 982–990. [DOI] [PubMed] [Google Scholar]
  • 162.Shemer I, Holmgren C, Min R, et al. Non-fibrillar beta-amyloid abates spike-timing-dependent synaptic potentiation at excitatory synapses in layer 2/3 of the neocortex by targeting postsynaptic AMPA receptors. Eur J Neurosci 2006; 23: 2035–2047. [DOI] [PubMed] [Google Scholar]
  • 163.Zhang Y, Guo O, Huo Y, et al. Amyloid-beta Induces AMPA Receptor Ubiquitination and Degradation in Primary Neurons and Human Brains of Alzheimer’s Disease. J Alzheimers Dis 2018; 62: 1789–1801. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Almeida CG, Tampellini D, Takahashi RH, et al. Beta-amyloid accumulation in APP mutant neurons reduces PSD-95 and GluR1 in synapses. Neurobiol Dis 2005; 20: 187–198. [DOI] [PubMed] [Google Scholar]
  • 165.Gu Z, Liu W and Yan Z. beta-Amyloid impairs AMPA receptor trafficking and function by reducing Ca2+/calmodulin-dependent protein kinase II synaptic distribution. J Biol Chem 2009; 284: 10639–10649. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Guntupalli S, Jang SE, Zhu T, et al. GluA1 subunit ubiquitination mediates amyloid-beta-induced loss of surface alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors. J Biol Chem 2017; 292: 8186–8194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.D’Amelio M, Cavallucci V, Middei S, et al. Caspase-3 triggers early synaptic dysfunction in a mouse model of Alzheimer’s disease. Nat Neurosci 2011; 14: 69–76. [DOI] [PubMed] [Google Scholar]
  • 168.Whitcomb DJ, Hogg EL, Regan P, et al. Intracellular oligomeric amyloid-beta rapidly regulates GluA1 subunit of AMPA receptor in the hippocampus. Sci Rep 2015; 5: 10934. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Bernareggi A, Duenas Z, Reyes-Ruiz JM, et al. Properties of glutamate receptors of Alzheimer’s disease brain transplanted to frog oocytes. Proc Natl Acad Sci U S A 2007; 104: 2956–2960. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Hsieh H, Boehm J, Sato C, et al. AMPAR removal underlies Abeta-induced synaptic depression and dendritic spine loss. Neuron 2006; 52: 831–843. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Reinders NR, Pao Y, Renner MC, et al. Amyloid-beta effects on synapses and memory require AMPA receptor subunit GluA3. Proc Natl Acad Sci U S A 2016; 113: E6526–E6534. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Fernandez-Perez EJ, Munoz B, Bascunan DA, et al. Synaptic dysregulation and hyperexcitability induced by intracellular amyloid beta oligomers. Aging Cell 2021; 20: e13455. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Hettinger JC, Lee H, Bu G, et al. AMPA-ergic regulation of amyloid-beta levels in an Alzheimer’s disease mouse model. Mol Neurodegener 2018; 13: 22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Piers TM, Kim DH, Kim BC, et al. Translational concepts of mGluR5 in synaptic diseases of the brain. Frontiers in pharmacology 2012; 3: 199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Um JW, Kaufman AC, Kostylev M, et al. Metabotropic glutamate receptor 5 is a coreceptor for Alzheimer abeta oligomer bound to cellular prion protein. Neuron 2013; 79: 887–902. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Hu NW, Nicoll AJ, Zhang D, et al. mGlu5 receptors and cellular prion protein mediate amyloid-beta-facilitated synaptic long-term depression in vivo. Nat Commun 2014; 5: 3374. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Haas LT, Salazar SV, Kostylev MA, et al. Metabotropic glutamate receptor 5 couples cellular prion protein to intracellular signalling in Alzheimer’s disease. Brain 2016; 139: 526–546. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Abd-Elrahman KS, Albaker A, de Souza JM, et al. Abeta oligomers induce pathophysiological mGluR5 signaling in Alzheimer’s disease model mice in a sex-selective manner. Sci Signal 2020; 13. [DOI] [PubMed] [Google Scholar]
  • 179.Renner M, Lacor PN, Velasco PT, et al. Deleterious effects of amyloid beta oligomers acting as an extracellular scaffold for mGluR5. Neuron 2010; 66: 739–754. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Wang Q, Walsh DM, Rowan MJ, et al. Block of long-term potentiation by naturally secreted and synthetic amyloid beta-peptide in hippocampal slices is mediated via activation of the kinases c-Jun N-terminal kinase, cyclin-dependent kinase 5, and p38 mitogen-activated protein kinase as well as metabotropic glutamate receptor type 5. J Neurosci 2004; 24: 3370–3378. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Hamilton A, Vasefi M, Vander Tuin C, et al. Chronic Pharmacological mGluR5 Inhibition Prevents Cognitive Impairment and Reduces Pathogenesis in an Alzheimer Disease Mouse Model. Cell Rep 2016; 15: 1859–1865. [DOI] [PubMed] [Google Scholar]
  • 182.Bellozi PMQ, Gomes GF, da Silva MCM, et al. A positive allosteric modulator of mGluR5 promotes neuroprotective effects in mouse models of Alzheimer’s disease. Neuropharmacology 2019; 160: 107785. [DOI] [PubMed] [Google Scholar]
  • 183.Everington EA, Gibbard AG, Swinny JD, et al. Molecular characterization of GABA-A receptor subunit diversity within major peripheral organs and their plasticity in response to early life psychosocial stress. Frontiers in Molecular Neuroscience 2018; 11: 18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Watanabe M, Maemura K, Kanbara K, et al. GABA and GABA receptors in the central nervous system and other organs. International review of cytology 2002; 213: 1–47. [DOI] [PubMed] [Google Scholar]
  • 185.Limon A, Reyes-Ruiz JM and Miledi R. Loss of functional GABA(A) receptors in the Alzheimer diseased brain. Proc Natl Acad Sci U S A 2012; 109: 10071–10076. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Li Y, Zhu K, Li N, et al. Reversible GABAergic dysfunction involved in hippocampal hyperactivity predicts early-stage Alzheimer disease in a mouse model. Alzheimers Res Ther 2021; 13: 114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Kelly L, Seifi M, Ma R, et al. Identification of intraneuronal amyloid beta oligomers in locus coeruleus neurons of Alzheimer’s patients and their potential impact on inhibitory neurotransmitter receptors and neuronal excitability. Neuropathol Appl Neurobiol 2021; 47: 488–505. [DOI] [PubMed] [Google Scholar]
  • 188.Calvo-Flores Guzman B, Kim S, Chawdhary B, et al. Amyloid-Beta(1–42) -Induced Increase in GABAergic Tonic Conductance in Mouse Hippocampal CA1 Pyramidal Cells. Molecules 2020; 25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Nava-Mesa MO, Jiménez-Díaz L, Yajeya J, et al. GABAergic neurotransmission and new strategies of neuromodulation to compensate synaptic dysfunction in early stages of Alzheimer’s disease. Front Cell Neurosci 2014; 8: 167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Lee BY, Ban JY and Seong YH. Chronic stimulation of GABAA receptor with muscimol reduces amyloid beta protein (25–35)-induced neurotoxicity in cultured rat cortical cells. Neurosci Res 2005; 52: 347–356. [DOI] [PubMed] [Google Scholar]
  • 191.Paula-Lima AC, De Felice FG, Brito-Moreira J, et al. Activation of GABA(A) receptors by taurine and muscimol blocks the neurotoxicity of beta-amyloid in rat hippocampal and cortical neurons. Neuropharmacology 2005; 49: 1140–1148. [DOI] [PubMed] [Google Scholar]
  • 192.Vinnakota C, Govindpani K, Tate WP, et al. An 5 GABAA Receptor Inverse Agonist, 5IA, Attenuates Amyloid Beta-Induced Neuronal Death in Mouse Hippocampal Cultures. Int J Mol Sci 2020; 21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Sawada M and Ichinose M. Amyloid beta proteins reduce the GABA-induced Cl- current in identified Aplysia neurons. Neurosci Lett 1996; 213: 213–215. [DOI] [PubMed] [Google Scholar]
  • 194.Yoshiike Y, Kimura T, Yamashita S, et al. GABA(A) receptor-mediated acceleration of aging-associated memory decline in APP/PS1 mice and its pharmacological treatment by picrotoxin. PLoS One 2008; 3: e3029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Sun B, Halabisky B, Zhou Y, et al. Imbalance between GABAergic and Glutamatergic Transmission Impairs Adult Neurogenesis in an Animal Model of Alzheimer’s Disease. Cell Stem Cell 2009; 5: 624–633. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Palop JJ, Chin J, Roberson ED, et al. Aberrant excitatory neuronal activity and compensatory remodeling of inhibitory hippocampal circuits in mouse models of Alzheimer’s disease. Neuron 2007; 55: 697–711. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Paula-Lima AC, Louzada PR, De Mello FG, et al. Neuroprotection against Abeta and glutamate toxicity by melatonin: are GABA receptors involved? Neurotox Res 2003; 5: 323–327. [DOI] [PubMed] [Google Scholar]
  • 198.Benarroch EE. GABAB receptors: structure, functions, and clinical implications. Neurology 2012; 78: 578–584. [DOI] [PubMed] [Google Scholar]
  • 199.Martin-Belmonte A, Aguado C, Alfaro-Ruiz R, et al. Density of GABA(B) Receptors Is Reduced in Granule Cells of the Hippocampus in a Mouse Model of Alzheimer’s Disease. Int J Mol Sci 2020; 21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Massone S, Vassallo I, Fiorino G, et al. 17A, a novel non-coding RNA, regulates GABA B alternative splicing and signaling in response to inflammatory stimuli and in Alzheimer disease. Neurobiol Dis 2011; 41: 308–317. [DOI] [PubMed] [Google Scholar]
  • 201.Kreis A, Desloovere J, Suelves N, et al. Overexpression of wild-type human amyloid precursor protein alters GABAergic transmission. Sci Rep 2021; 11: 17600. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Almasi A, Zarei M, Raoufi S, et al. Influence of hippocampal GABA(B) receptor inhibition on memory in rats with acute beta-amyloid toxicity. Metab Brain Dis 2018; 33: 1859–1867. [DOI] [PubMed] [Google Scholar]
  • 203.Rodenkirch C, Carmel JB and Wang Q. Rapid Effects of Vagus Nerve Stimulation on Sensory Processing Through Activation of Neuromodulatory Systems. Frontiers in Neuroscience 2022; 16. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

This study did not involve the collection or generation of new data.

RESOURCES