Your new experience awaits. Try the new design now and help us make it even better

REVIEW article

Front. Aging Neurosci., 24 July 2025

Sec. Cellular and Molecular Mechanisms of Brain-aging

Volume 17 - 2025 | https://doi.org/10.3389/fnagi.2025.1604378

NMDA receptors in neurodegenerative diseases: mechanisms and emerging therapeutic strategies

Keyi Zhang,&#x;Keyi Zhang1,2Ming Wen,&#x;Ming Wen1,3Xinyue Nan,Xinyue Nan1,2Shuaizhu Zhao,Shuaizhu Zhao1,2Hao Li,
Hao Li1,2*Yanping Ai,
Yanping Ai1,4*Houze Zhu,
Houze Zhu1,5*
  • 1Innovation Center for Brain Medical Sciences, The Ministry of Education of the People’s Republic of China, Huazhong University of Science and Technology, Wuhan, China
  • 2Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
  • 3Department of Neurosurgery, Wuhan Hankou Hospital, Hankou Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
  • 4Department of Neurology, Wuhan Hankou Hospital, Wuhan, China
  • 5Department of Physiology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, China

NMDA receptors (NMDARs) are widely distributed throughout the central nervous system (CNS) and play pivotal roles in normal physiological processes such as synaptic plasticity, learning, and memory. Substantial evidence indicates that NMDAR dysfunction, particularly excessive calcium influx, critically contributes to the pathogenesis of major neurodegenerative diseases, including Alzheimer’s disease (AD), Parkinson’s disease (PD), Huntington’s disease (HD), and amyotrophic lateral sclerosis (ALS). Dysregulated glutamatergic signaling synergizes with pathological protein aggregation (e.g., Aβ, α-synuclein, mutant huntingtin) to drive neuronal loss. We systematically delineate NMDAR-related mechanisms underlying neurodegeneration, highlighting spatial-specific roles (e.g., synaptic NMDAR-mediated neuroprotection versus extrasynaptic NMDAR-mediated excitotoxicity) and crosstalk with mitochondrial dysfunction and oxidative stress. We critically evaluate current therapeutic strategies targeting NMDARs, including subunit-selective modulators, downstream effector modulation, and glutamate transporter modulation designed to restore NMDAR homeostasis. Consequently, NMDARs and their modulators represent promising therapeutic targets for these refractory conditions. This review comprehensively summarizes current research on the involvement of NMDARs and the glutamatergic system in neurodegenerative diseases. Furthermore, we discuss the clinical application of NMDAR-targeting agents and explore emerging therapeutic strategies focused on modulating NMDAR-related pathways. This article aims to provide a reference for elucidating the molecular mechanisms underlying these neurodegenerative disorders and to highlight potential avenues for future drug development.

1 Introduction

Glutamatergic neurotransmission, primarily mediated by N-methyl-D-aspartate receptors (NMDARs), underpins synaptic plasticity, learning, memory and other critical physiological functions (Bannerman et al., 2014; Paoletti et al., 2013; Morris, 2013). However, this critical signaling pathway exhibits a profound duality: its exquisite calcium permeability, essential for physiological processes like long-term potentiation (LTP) (Lüscher and Malenka, 2012), concurrently renders neurons vulnerable to pathological cascades (Kodis et al., 2018). Crucially, NMDAR dysregulation is now recognized not merely as a consequence but as a central driver of the progressive neuronal dysfunction and loss in major neurodegerative diseases, including amyotrophic lateral sclerosis (ALS) (Paul and de Belleroche, 2014; Spalloni et al., 2013), Parkinson’s disease (PD) (Xu et al., 2012; Picconi et al., 2012), Huntington’s disease (HD) (Sepers and Raymond, 2014; Fernandes and Raymond, 2009) and Alzheimer’s disease (AD) (Xu et al., 2012; Wang and Reddy, 2017; Babaei, 2021).

While the neurotoxic potential of excessive NMDAR activation, termed “excitotoxicity,” has been a milestone concept since its first description by Olney in the 1970s (Rothman and Olney, 1987), contemporary research is rapidly dismantling simplistic views, revealing context-dependent signaling outcomes. This complexity simultaneously illuminates novel therapeutic avenues.

The frontier of neurodegeneration research has moved decisively beyond a monolithic view of the NMDARs. Key advances have demonstrated that functional consequences hinge on dynamic interactions among: subunit composition (GluN2A versus GluN2B), subcellular localization (synaptic versus extrasynaptic), developmental stage, neuronal subtype, and associated proteins including scaffolding proteins and signaling effectors (Paoletti et al., 2013; Lohmann and Kessels, 2014; Gladding and Raymond, 2011; Sanz-Clemente et al., 2013). The oversimplified dichotomy attributing neuroprotection exclusively to GluN2A-containing receptors and toxicity to GluN2B-containing receptors has evolved into a more nuanced understanding paradigm. No NMDAR subunit is intrinsically “good” or “bad”; their roles are context-dependent (Hardingham and Bading, 2003). Synaptic NMDARs – often enriched in GluN2A subunits in mature neurons, typically activate pro-survival pathways supporting neuronal plasticity and survival. In contrast, extrasynaptic pools, which frequently contain GluN2B, preferentially couple to mitochondrial dysfunction and oxidative stress when chronically overactivated (such as pathological glutamate spillover) (Hardingham and Bading, 2010). Subunits with restricted expression (such as GluN2D and GluN3A) contribute uniquely to disease-specific vulnerabilities in AD and PD (Crawley et al., 2022; Mellone et al., 2019; Swanger et al., 2015). Developmental and pathological reprogramming further complicates this landscape: GluN2B dominance is vital in neurodevelopment but exacerbates excitotoxicity in mature degenerating neurons. Conversely, AD induces pathological GluN2A internalization and GluN2B surface accumulation, favoring excitotoxic signaling.

Pathologically, neurodegenerative processes actively corrupt this finely tuned NMDAR signaling system. Several universal mechanisms are shared by different neurodegenerative diseases by which NMDAR dysfunction propagates neurodegeneration: (1) excitotoxic calcium overload (Dong et al., 2009; Lau and Tymianski, 2010); (2) synaptic/extrasynaptic receptor imbalance (Hardingham and Bading, 2010; Parsons and Raymond, 2014), (3) mitochondrial dysfunction and oxidative stress (Lin and Beal, 2006; Islam, 2017), and (4) proteinopathy-induced receptor mislocalization (Aβ in AD; α-synuclein in PD) (Wang et al., 2013; Durante et al., 2019). Understanding these convergent pathways is key to developing broad-spectrum therapeutic strategies.

The limited disease-modifying efficacy of broad antagonists such as memantine (an uncompetitive, low-affinity, open-channel blocker that preferentially blocks the extrasynaptic NMDAR) (Johnson and Kotermanski, 2006) underscores the futility of non-selective blockade and the imperative for precision targeting. Current strategies focus on: subunit-selective modulators, including GluN2B-selective negative modulators (such as ifenprodil and its derivatives) (Egunlusi and Joubert, 2024) or GluN2A-selective positive modulators (which are less investigated) (Hanson et al., 2020), aiming to restore subunit balance. Location-biased interventions strive to selectively inhibit pathological extrasynaptic NMDAR signaling while sparing crucial synaptic function. Novel strategies such as modulation of NMDAR upstream kinases and phosphatases (such as PKC) and downstream effectors (such as DAPK), dissociation of NMDA receptor complex are also emerging and under active investigation.

This review synthesizes the latest mechanistic insights into multifaceted NMDAR dysregulation in neurodegeneration. We critically evaluate subunit-specific, localization-dependent, and context-governed signaling in disease models and human pathology. Emphasis is placed on translating this complexity into emerging therapeutics that restore physiological homeostasis or selectively block pathological cascades, moving beyond crude receptor inhibition.

2 NMDAR composition and spatial distribution

Structurally, functional NMDARs are obligate heterotetramers, typically assembled as dimeric pairs of glycine-binding GluN1 subunits (the obligatory co-agonist site) and glutamate-binding GluN2 subunits (the agonist site) (Cull-Candy et al., 2001; Hansen et al., 2018) (Figure 1). The GluN1 subunit (encoded by GRIN1) is essential for channel activity and is ubiquitous in all NMDARs (Tu and Kuo, 2015; Chou et al., 2024). The critical functional diversity is conferred primarily from the incorporation of GluN2 subunits (GluN2A-D, encoded by GRIN2A-D, respectively), each conferring distinct biophysical, pharmacological, and signaling properties to the receptor complex (Wyllie et al., 2013; Paoletti, 2011). GluN2A and GluN2B predominate in the forebrain, particularly in specific regions such as the hippocampus and cortex. GluN2C and GluN2D are more restricted, expressed notably in the cerebellum, thalamus, and during early development (Monyer et al., 1994; Watanabe et al., 1994; Köhr, 2006). Additionally, GluN3 subunits (GluN3A-B, encoded by GRIN3A-B), which are not a component of most natural NMDARs, can incorporate into complexes alongside GluN1 and GluN2, forming non-canonical glycine-activated receptors characterized by low calcium permeability and insensitive to magnesium block (Henson et al., 2010; Chatterton et al., 2002). This combinatorial assembly generates a vast array of receptor subtypes with tailored properties.

Figure 1
Diagram of NMDAR subunit composition and activity. The GluN1 subunit binds with glycine or D-serine, and it is essential for NMDAR channel activity. The GluN2 subunit binds with glutamate, determining NMDAR channel properties. The exact function of GluN3 remains unclear but can assemble with other GluN1 or GluN2 subunits to form atypical NMDARs with distinct properties.

Figure 1. Subunit composition of NMDA receptors. This schematic illustrates the heterotetrameric organization of the NMDARs. Structurally, functional NMDARs are obligate heterotetramers, typically assembled as dimeric pairs of GluN1 subunits and GluN2 subunits. The GluN1 subunit is essential for channel activity, binds the coagonists glycine or D-serine, and exhibits functional diversity through alternative splicing. The regulatory GluN2 subunit, which binds the primary agonist glutamate, confers distinct biophysical properties, pharmacological profiles, and signaling capabilities to the receptor complex. While the precise roles of GluN3 subunits are less defined, emerging evidence indicates their incorporation forms non-canonical NMDARs characterized by reduced calcium permeability and insensitivity to voltage-dependent magnesium block, suggesting unique contributions to neuropathological mechanisms and potential therapeutic targeting.

The spatial distribution of NMDARs within neurons is highly organized and functionally critical. Crucially, NMDARs exhibit a distinct synaptic versus extrasynaptic localization, a dichotomy with profound implications for neuronal signaling and survival (Gladding and Raymond, 2011; Parsons and Raymond, 2014). Synaptic NMDARs, anchored by scaffolding proteins like the PSD-95 family to the postsynaptic density (PSD) (Chen et al., 2015; Niethammer et al., 1996), are activated by vesicular glutamate release into the synaptic cleft. They are key mediators of physiological processes, including the induction of long-term potentiation (LTP) and long-term depression (LTD), the foundation of learning and memory (Lüscher and Malenka, 2012; Bliss and Collingridge, 1993). In contrast, extrasynaptic NMDARs, localized to the plasma membrane outside the PSD such as the dendritic shaft and soma, are often associated with different scaffolds proteins (SAP102 and SAP97) or adhesion molecules (cadherin and catenin) (Petralia et al., 2010; Groc et al., 2009; Li et al., 2011), are primarily activated under pathological conditions involving excessive glutamate spillover or impaired astrocytic glutamate reuptake.

Critically, the location of NMDAR also determines the downstream transcriptional consequences (Bading, 2017). Synaptic NMDAR activation promotes phosphorylation of the transcription factor cAMP response element-binding protein (CREB), driving expression of pro-survival, anti-apoptotic genes such as the brain-derived neurotrophic factor (BDNF) (Greer and Greenberg, 2008) Conversely, extrasynaptic NMDARs induce CREB shut-off pathway while concurrently activating pro-death signaling pathways (Hardingham et al., 2002).

Collectively, this intricate molecular architecture, defined by subunit composition, post-translational modifications, and precise subcellular positioning, establishes the NMDAR as a pivotal regulator of normal brain function. Consequently, disruptions in subunit expression, trafficking, synaptic localization, or downstream signaling cascades contribute profoundly to the pathogenesis of diverse neurodegenerative diseases.

3 NMDAR in neuronal impairment: excitotoxicity and beyond

3.1 NMDAR-induced excitotoxicity

Neuronal responses to NMDA receptor activity follow a bell-shaped curve where neuronal survival peaks at physiological activation levels but declines under both hypoactive and hyperactive states (Hardingham and Bading, 2010). Hypoactivation of NMDAR has been recognized as a crucial driver in the progression and manifestation of age-related cognitive decline through inhibiting NMDAR-induced LTP and synaptic plasticity and other diseases such as schizophrenia (Nakazawa and Sapkota, 2020; Lindsley et al., 2006; Dong et al., 2023). This paradigm, first established by Hardingham and Bading, arises from spatiotemporal segregation of receptor subtypes. Under physiological conditions, NMDA receptors are briefly activated by a saturating (~1 mM) concentration of glutamate to conduct synaptic transmission (Vyklicky et al., 2014). While under conditions of hypoactivation, NMDAR hypofunction leads to drastic alterations in calcium influx and cellular signalling, impairing receptor transport to the postsynaptic membrane. Conversely, hyperactivation, caused by pathological glutamate spillover (μM concentrations of glutamate) according to different types of pathology or impaired receptor internalization, triggers downstream excitotoxic cascades including ROS production, mitochrondrial dysfunction and eventually neuronal death (Papadia et al., 2005).

Excitotoxicity, first described by Olney in the 1970s (Rothman and Olney, 1987), is a critical neurodegenerative mechanism wherein excessive NMDAR activation triggers neuronal death through calcium overload and downstream cytotoxic cascades (Rothman and Olney, 1995) (see Figure 2). This cascade initiates with disrupted glutamate homeostasis. Under physiological conditions, extracellular glutamate concentrations are regulated by excitatory amino acid transporters (EAATs), predominantly EAAT1/2 on astrocytes. The glutamate reuptake mechanism of EAATs fundamentally relies on the transmembrane sodium gradient established and maintained by ATP-fueled Na+/K+-ATPase activity (Alleva et al., 2020; Alleva et al., 2022). This indirect but absolute energy dependence creates a critical vulnerability point: when under conditions of ischemia, mitochondrial dysfunction, or oxidative stress, which impair ATP production and inhibit the normal function of ion pumps, can lead to the collapse of the ion gradient. Consequently, EAAT-mediated glutamate clearance fails, allowing glutamate to persistently activate postsynaptic NMDARs (Andersen et al., 2021; Murphy-Royal et al., 2017; Mahmoud et al., 2019).

Figure 2
Diagram depicting NMDAR-mediated excitotoxic cascade. Pathological conditions may impair ATP production, inihibit ion pump activitiy and can lead to the collapse of the ion gradient, which eventually lead to abnormal glutamate level in the synaptic cleft. Persistent glutamate accumulation can lead to sustained NMDAR overactivation, triggering calcium-dependent neurotoxic cascades, including further mitochondrial dysfunction and neuronal death.

Figure 2. NMDAR-mediated excitotoxic cascade. This schematic illustrates the core sequence of molecular events driving glutamate excitotoxicity: The glutamate reuptake by EAATs fundamentally relies on the transmembrane sodium gradient established and maintained by ATP-dependent Na+/K+-ATPase activity. Pathological conditions of ischemia, mitochondrial dysfunction, or oxidative stress, impair ATP production, inhibit ion pumps, can lead to the collapse of the ion gradient. Consequently, astrocytic EAATs fail to clear synaptic glutamate. Persistent glutamate accumulation caused sustained NMDAR overactivation, triggering pathological calcium influx that initiates downstream neurotoxic cascades, including further mitochondrial dysfunction and neuronal death.

Overactivation of NMDARs further promote calcium influx, initiating downstream neurotoxic cascades: Intracellular calcium surge overwhelms mitochondrial buffering capacity, inducing mitochondrial membrane depolarization, halting ATP synthesis, and reactive oxygen species (ROS) explosion (Neves et al., 2023; Rego and Oliveira, 2003; Szydlowska and Tymianski, 2010). Concurrently, calcium-dependent enzymes initiate destructive processes: calpains degrade cytoskeletal proteins and activate pro-apoptotic Bcl-2 family members (Raynaud and Marcilhac, 2006; Chan and Mattson, 1999); NO causes inhibition of mitochondrial respiratory chain, rapid glutamate release from both astrocytes and neurons, and subsequent excitotoxic death of the neurons (Dawson et al., 1991; Brown and Bal-Price, 2003); phospholipase A2 (PLA2) promotes arachidonic acid (AA) release, fueling inflammatory cascades (Sun et al., 2004).

These pathways converge via necrosis, apoptosis, or dysregulated autophagy, constituting a final common pathway in neurodegeneration.

3.2 Beyond excitotoxicity: NMDAR as an amplifier of neuronal damage

While the etiology of neurodegenerative diseases remains incompletely understood, emerging evidence implicates defects in energy metabolism and oxidative damage as key co-pathogenic mechanisms beyond excitotoxicity (Beal, 1995; Tripathi et al., 2020; Stark and Bazan, 2011). Involvement of oxidative damage and mitochondrial dysfunction has been suggested as a common feature shared by multiple neurodegenerative diseases (Cenini et al., 2019; Reed, 2011).

Oxidative stress is suggested to be involved in the etiology of both brain aging and neurodegenerative diseases such as AD and PD. (Puspita et al., 2017; Malinski, 2007) NMDAR overactivation leads to excessive calcium influx, which not only triggers excitotoxic cell death but also initiates a cascade of oxidative events. Elevated intracellular calcium binds with calmodulin and activates neuronal nitric oxide synthase (nNOS), to convert l-arginine to citrulline and nitric oxide (NO). NO can function as a messenger molecule in the CNS under physiological conditions, however, when generated in excessive amounts, NO can be neurotoxic (Dawson and Dawson, 1996). NO can be scavenged in a rapid reaction with superoxide (O2) to generate peroxynitrite (ONOO). ONOO is a potent oxidant and the primary component of nitroxidative stress (Malinski, 2007). These free radicals further impair mitochondrial function, creating a vicious cycle of oxidative damage (Duncan and Heales, 2005; Ghasemi et al., 2018). Notably, mitochondrial dysfunction exacerbates ROS production (Murphy, 2009), which may establish a feedforward loop that accelerates neurodegeneration.

Lipid peroxidation, as a critical downstream consequence, directly links oxidative stress to NMDAR regulation and participates in AD, HD, and PD pathophysiology (Reed, 2011). ROS attack polyunsaturated fatty acids (PUFAs) in neuronal membranes to generate highly reactive aldehydes such as 4-hydroxynonenal (4-HNE) and malondialdehyde (MDA) (Gaschler and Stockwell, 2017). These lipid peroxidation products can impair glutamate transport through HHE modification of EAAT2 (Lovell et al., 2012), reducing glutamate clearance and further potentiating NMDAR overactivation.

4 Flux-independent NMDAR signaling

Beyond its canonical role as an ionotropic receptor mediating fast excitatory synaptic transmission, emerging evidence highlights the significance of flux-independent (non-canonical or metabotropic-like) NMDAR signaling in neurodegenerative diseases. Several studies have indicated that flux-independent NMDARs mediate LTD, cell membrane molecular dynamics, pH sensing, and synaptic depression induced by amyloid-β (Aβ) oligomers (de Oca and Balderas, 2018; Park et al., 2022). Tamburri et al. (2013) demonstrated that oligomeric Aβ induces rapid synaptic depression in hippocampal neurons of slices through a mechanism independent of ion influx but dependent on synaptic NMDAR activaion. Kessels et al. further established that Aβ-induced synaptic depression requires GluN2B-containing NMDARs, as evidenced by blockade with the competitive antagonist D-2-Amino-5-phosphonopentanoic acid (D-APV) (a non-selective GluN2 antagonist) but not by the open-channel blocker MK-801 or glycine-site antagonist 7 chloro-kynurenate (7CK) (Kessels et al., 2013). However, Nabavi et al. reported a contradictory result that 7CK failed to block LTD, which was suspected to be caused by subtle methodological difference (Nabavi et al., 2014). Collectively, these findings support the involvement of flux-independent NMDAR signaling in synaptic dysregulation during neurodegeneration. While this emerging paradigm holds significant therapeutic promise, important limitations remain: the precise molecular mechanisms underlying flux-independent NMDAR signaling are still incompletely characterized, and like most new knowledge, flux-independent NMDARs has been controversial, as contradictory findings exist. These questions are currently under active investigation.

5 NMDAR in neurodegenerative diseases

5.1 NMDAR and amyotrophic lateral sclerosis (ALS)

Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disease characterized by the progressive loss of upper and lower motor neurons, leading to muscle atrophy, paralysis, and ultimately respiratory failure (Hardiman et al., 2017; Feldman et al., 2022). Although its etiology remains incompletely defined, growing evidence implicates glutamate-mediated excitotoxicity as a key contributor to motor neuron degeneration (Paul and de Belleroche, 2014; Spalloni et al., 2013; Heath and Shaw, 2002).

Motor neurons are particularly vulnerable to NMDAR-mediated excitotoxicity due to their low calcium-buffering capacity and abundant NMDAR expression (Van Den Bosch et al., 2006). Preclinical studies and patient-derived motor neurons have demonstrated that NMDAR overactivation results in mitochondrial dysfunction, oxidative stress, and activation of apoptotic pathways, ultimately culminating in motor neuron death (Paul and de Belleroche, 2014; Heath and Shaw, 2002; Menzies et al., 2002; Catania et al., 2001; Paul et al., 2014).

Interrupted glutamate homeostasis further contributes to excitotoxicity. Studies have revealed elevated levels of glutamate and aspartate in the cerebrospinal fluid of ALS patients (Rothstein et al., 1990). This accumulation is strongly linked to the selective loss or dysfunction of the major astrocytic glutamate transporter, EAAT2, observed in the motor cortex and spinal cord of ALS patients and in transgenic ALS mouse (SOD1 mutant) models (Rothstein et al., 1995; Howland et al., 2002). Critically, the reduced EAAT2 expression is induced by dysregulated NF-κB signaling, which represses EAAT2 expression (Frakes et al., 2014; Crosio et al., 2011). Paradoxically, NF-κB is also required for both activation and repression of the EAAT2 promoter, which positioning it as a context-dependent regulator (Kim et al., 2011). For example, N-myc and NF-κB are required for TNF-α-mediated transcriptional repression of EAAT2. On the contrary, NF-κB also mediates EGF-, TGF-α-, and cAMP-induced EAAT2 promoter activation (Su et al., 2003; Sitcheran et al., 2005). Besides its effects on glutamate transporters, NF-κB may also contribute to ALS pathogenesis by induction of pro-inflammatory gene expression (Källstig et al., 2021). Consequently, excessive synaptic glutamate persistently activates NMDARs. Oxidative stress may also participate in ALS pathogenesis through oxidative stress-mediated protein injury, lipid peroxidation, and DNA and RNA oxidation have been observed in ALS patients (Singh et al., 2019).

Beyond glutamate, pathological accumulation of the NMDAR co-agonist D-serine driven by impaired degradation such as the DAO R199W mutation shifts its role from physiological modulator to neurotoxic effector (Sasabe et al., 2012). Excess D-serine drives NMDAR overstimulation that exacerbates neurodegeneration through two convergent pathways: direct potentiation of excitotoxic calcium influx via extrasynaptic NMDARs, and indirect induction of non-excitotoxic death mechanisms including autophagic flux blockade and intrinsic apoptosis (Mitchell et al., 2010; Kondori et al., 2018).

ALS-associated genetic mutations further implicate NMDAR dysfunction. For example, the C9ORF72 mutation, the most common genetic cause of ALS/FTD (Pang and Hu, 2021), contributes to neurodegeneration through disrupting the surface expression, transport, and recycling of NMDARs. Studies using induced motor neurons (iMNs) derived from C9ORF72 ALS/FTD patients revealed elevated expression of the essential GluN1 subunit on neurites and dendritic spines (Burk and Pasterkamp, 2019; Shi et al., 2018). This increase facilitates more frequent calcium influx, thereby exacerbating excitotoxicity.

The central role of NMDAR-mediated excitotoxicity makes NMDAR a compelling therapeutic target in ALS. However, the complexity of ALS pathogenesis (Figure 3) has hindered the development of effective disease-modifying treatments. The only FDA-approved drug, riluzole, has modest efficacy, and can only extend the average survival time by 3 months and cannot reverse motor neuron damage (Miller et al., 2012). Direct NMDAR antagonists (such as memantine) have yielded limited clinical success, largely due to their disruptive effects on essential physiological NMDAR functions in synaptic plasticity and cognition, leading to unacceptable side effects. While preclinical studies in SOD1 mutant mice demonstrate that memantine treatment delays disease progression and improves motor neuron survival, likely through inhibition of spinal cord NMDA receptors (Wang and Zhang, 2005; Joo et al., 2007), these findings have not translated to clinical benefit in ALS patients. Notably, several clinical trials evaluating memantine in sporadic ALS showed that though memantine is well-tolerated in ALS patients, no significant effects on disease progression or survival time were found (de Carvalho et al., 2010; Bhai et al., 2025). Thus, while memantine remains a valuable tool for investigating NMDAR-mediated mechanisms in ALS models, current evidence does not support its therapeutic use in patients.

Figure 3
Proposed text: Diagram illustrating the key mechanisms driving excitotoxicity in ALS. Pathologically elevated levels of D-serine in ALS resulting in NMDAR overactivation. Altered NMDAR trafficking and increased surface expression may also contribute to this overactivation, facilitating pathological calcium influx and exacerbating excitotoxic injury to motor neurons.

Figure 3. NMDAR dysregulation in amyotrophic lateral sclerosis (ALS). This schematic illustrates the key mechanisms driving excitotoxicity in ALS. Interrupted glutamate homeostasis is a key promoter in the pathogenesis of ALS, primarily due to selective loss or dysfunction of EAAT2. Pathologically elevated levels of the NMDAR coagonist D-serine, resulting from impaired degradation due to the DAO R199W mutation, further promote NMDAR activation. Altered NMDAR trafficking and increased surface expression, characterized by elevated GluN1 subunit density in neurons harboring C9ORF72 mutations, provide an additional substrate for overactivation. These astrocytic and neuronal defects collectively sustain NMDAR overactivation, facilitating pathological calcium influx and exacerbating excitotoxic injury to motor neurons.

Consequently, research is shifting towards more refined approaches, such as subunit-selective NMDAR antagonists or modulators targeting allosteric sites. Additionally, combination therapies targeting both upstream triggers (such as enhancing glutamate uptake) or downstream effectors of NMDAR overactivation (such as anti-apoptotic agents, antioxidants), even gene therapies, alongside selective NMDAR modulation hold promise for providing broader neuroprotection for ALS patients (Jiang et al., 2022).

5.2 NMDAR and Parkinson’s disease (PD)

Parkinson’s disease (PD) is a progressive neurodegenerative disorder characterized by the loss of dopaminergic neurons in the substantia nigra pars compacta (SNc) and the formation of Lewy bodies. This pathology leads to core motor symptoms including bradykinesia, rigidity, and resting tremor, as well as non-motor manifestations such as cognitive impairment and psychiatric disturbances (Kalia and Lang, 2015; Hayes, 2019; Melzer and Monyer, 2020).

Chronic NMDAR overactivation is a key mechanism of dopaminergic neuron degeneration (Beal, 1998). Basal ganglia circuit imbalance, specifically disinhibition of the subthalamic nucleus (STN) due to striatal dopamine depletion, drives excessive glutamate release onto substantia nigra pars compacta (SNc) neurons, resulting in sustained NMDAR stimulation and downstream neurotoxic events (Dunah et al., 2000; Rodriguez et al., 1998). This excitotoxic cascade is compounded by two intrinsic vulnerabilities of SNc neurons: low expression of calcium-buffering proteins (notably calbindin-D28K) (Liang et al., 1996; Sulzer and Surmeier, 2013) and high surface density of GluN2B-containing NMDARs (Suárez et al., 2010; Jones and Gibb, 2005). Besides, misfolded α-synuclein oligomers exacerbate NMDAR hyperactivation by inducing astrocytic glutamate release (Trudler et al., 2021). These interactions jointly trap NMDARs in a hyperactive state, exacerbating calcium influx and overwhelming mitochondrial calcium buffering capacity.

Beyond excitotoxicity, NMDAR dysregulation in PD also contributes to impaired synaptic plasticity and the non-motor symptoms of the disease, such as cognitive impairment and depression. An unbalanced GluN2A/GluN2B subunit ratio of the striatal synaptic NMDAR is thought to be a crucial determinant in the regulation of motor behaviour and synaptic plasticity in PD (Mellone et al., 2015). Pathological alterations include: (1) an imbalanced GluN2A/GluN2B ratio with selective depletion of GluN2B-containing receptors; (2) reduced phosphorylation of GluN1 and GluN2B; (3) dopamine D1 receptor-dependent redistribution of NMDARs between synaptic and postsynaptic sites (Landwehrmeyer et al., 1995; Paillé et al., 2010; Zhang and Chergui, 2015; Dunah and Standaert, 2001). These changes collectively impair synaptic function and may mediate adverse effects of dopaminergic therapy such as the levodopa-induced dyskinesia (LID) (Zhang et al., 2023).

Therapeutically, NMDAR antagonists show dual promise: they protect SNc neurons in preclinical models and ameliorate motor complications. Amantadine, an antagonist of NMDAR as an adjuvant to levodopa therapy, has been found to significantly ameliorate motor complications in PD and supports the idea that NMDAR hyperfunction contributes to levodopa-associated motor complications (Papa and Chase, 1996; Greenamyre and O'Brien, 1991; Metman et al., 1998). Memantine exhibits more modest efficacy and lacks amatadine’s anti-dyskinetic activity (Merello et al., 1999). The second generation of adamantane-based drugs is being designed, seeking to improve the clinical efficacy (Dembitsky et al., 2020). Given the dual role of NMDARs in motor and non-motor symptoms, future therapies may need to adopt a multifaceted approach, targeting specific receptor subtypes or brain regions to address the diverse manifestations of PD.

5.3 NMDAR and Huntington’s disease (HD)

Huntington’s disease (HD) is an autosomal dominant disorder caused by CAG trinucleotide expansions in the HTT gene that results in polyglutamine (polyQ)-expanded mutant huntingtin (mHTT) protein. This mutation drives progressive striatal degeneration largely through mHTT-induced NMDAR dysregulation, manifesting as motor dysfunction, cognitive decline, and psychiatric disturbances (McColgan and Tabrizi, 2018; Walker, 2007).

The selective vulnerability of striatal medium spiny neurons (MSNs) stems from their high GluN2B-NMDAR expression and intense corticostriatal glutamatergic input, rendering them particularly sensitive to mHTT-induced alterations in NMDAR trafficking, localization, and signaling (Landwehrmeyer et al., 1995; Li et al., 2003; Cepeda et al., 2007).

The pathogenic cascade begins with mHTT disrupting postsynaptic organization: mHTT exhibits reduced binding to PSD-95 compared to wild-type HTT (Sun et al., 2001). Yet mHTT paradoxically enhances PSD-95/GluN2B interactions in HD models, which may be linked to increased extrasynaptic NMDAR mislocation in HD (Fan et al., 2009; Milnerwood et al., 2010). This shift toward extrasynaptic NMDAR dominance creates a permissive environment for excitotoxicity, which has been suggested as a major player in HD pathogenesis (Fan and Raymond, 2007; Raymond et al., 2011).

Concurrently, elevated GluN3A subunit expression in HD striatum accelerates afferent synapse loss onto medium spiny neurons (MSNs). Notably, suppressing GluN3A in the YAC128 HD mouse model corrects NMDAR hyperexcitability, rescues synapses, ameliorates motor and cognitive deficits, and reduces striatal atrophy (Marco et al., 2013; Wesseling and Pérez-Otaño, 2015).

Therapeutic strategies targeting NMDAR dysfunction in HD are evolving to address the complexity of receptor dysregulation, with a growing emphasis on subunit-selective modulation and the restoration of synaptic-extrasynaptic NMDAR balance. GluN2B-selective antagonists such as memantine have shown considerable promise in preclinical models. In a small pilot trial in HD patients, 20 mg of memantine daily intake can significantly improve motor symptoms (Ondo et al., 2007). Besides, the application of neurotrophic factors, autophagy regulators, stem cells, and genetic therapies are also under investigation for HD treatment (Kim et al., 2021).

5.4 NMDAR and Alzheimer’s disease (AD)

Alzheimer’s disease (AD), the most prevalent neurodegenerative cause of dementia, is defined by progressive cognitive decline alongside neuropathological hallmarks including amyloid-β (Aβ) plaques and neurofibrillary tau tangles (Lane et al., 2018; Scheltens et al., 2021). Central to AD pathogenesis is NMDAR dysregulation, manifesting through excitotoxicity, synaptic failure, and bidirectional interactions with Aβ/tau pathology (Wang and Reddy, 2017; Raïch et al., 2024; Escamilla et al., 2024) (see Figure 4).

Figure 4
Diagram illustrating Aβ-triggered pathological cycle in AD. Aβ oligomers drive presynaptic glutamate spillover through multiple pathways. Postsynaptic NMDARs undergo Aβ-induced phosphorylation and internalization, leading to pathological overactivation and calcium overload. Resultant calcium overload triggers mitochondrial dysfunction, ROS production, energy failure and eventually neuronal death.

Figure 4. Aβ-induced excitotoxic cascade via NMDAR dysregulation in Alzheimer’s disease. This schematic illustrates the Aβ-triggered pathological cycle: Aβ oligomers drive presynaptic glutamate spillover through impaired glutamate transporter expression, decreased glutamine synthetase activity, and altered vesicular glutamate release. Postsynaptic NMDARs undergo Aβ-induced phosphorylation and internalization, leading to pathological overactivation and calcium influx dysregulation. Resultant calcium overload triggers mitochondrial dysfunction, reactive oxygen species (ROS) production, energy failure and eventually neuronal death. This self-amplifying cascade culminates in neuronal edema, synaptic loss, and neurodegeneration.

Aβ oligomers were described to accumulate in the AD patient brain, or in vitro in human cortex neuronal cultures, at GluN2B-containing synapses. Aβ oligomers bind to cellular prion protein (PrPc) and metabotropic glutamate receptor 5 (mGluR5), forming a complex that promotes phosphorylation of the GluN2B subunits. This triggers initial NMDAR surface accumulation followed by internalization, driving calcium overload, mitochondrial dysfunction, calpain activation, and dendritic spine loss (Um et al., 2013; Um et al., 2012; De Felice et al., 2007). Concurrently, Aβ disrupts glutamate homeostasis by mislocalizing astrocytic EAAT2 transporters and impairing glutamine synthetase (GS) activity, elevating extracellular glutamate and sustaining NMDAR stimulation (Scimemi et al., 2013; Aksenov et al., 1995). Aβ additionally induces pathological NMDAR subunit switching (GluN2B to GluN2A) and alters vesicular glutamate release via vGluT1 and/or vGluT2 downregulation (Kessels et al., 2013; Rodriguez-Perdigon et al., 2016; Mi et al., 2023). Additionally, reduced activity and expression of GS have been reported in both patient tissue and animal models, contributing to the glutamate homeostasis disruption (Kulijewicz-Nawrot et al., 2013; Robinson, 2000). Tau pathology amplifies this dysfunction when hyperphosphorylated tau impairs synaptic NMDAR trafficking while promoting receptor internalization (Hoover et al., 2010).

The distribution of synaptic versus extrasynaptic NMDARs has emerged as a key profile in neurodegenerative diseases including AD. In AD, extrasynaptic NMDARs oppose synaptic NMDARs by triggering CREB (a master regulator of synaptic plasticity) shut-off and promoting mitochondrial dysfunction and neuronal death (Hardingham et al., 2002; Wang et al., 2004; Esposito et al., 2013). CREB is a crucial molecular factor for learning and memory and its downregulation is assumed to result in cognitive deficits in AD (Rosa and Fahnestock, 2015; Bartolotti et al., 2016). Critically, Aβ downregulates CREB phosphorylation, and suppresses downstream BDNF expression (Amidfar et al., 2020; Garzon and Fahnestock, 2007).

Mitochondrial-ROS dysregulation converges on NMDAR pathology: Reduced PGC-1α levels in AD impair ROS detoxification and reduce mitochondrial density, diminishing neuronal resilience to excitotoxic stress (Sweeney and Song, 2016; Qin et al., 2009; Wareski et al., 2009; Cui et al., 2006).

Mechanistically, NF-κB signaling also plays a crucial role in AD pathogenesis by regulating different molecules responsible for promoting the morbidities associated with AD (Sun et al., 2022). NF-κB induces the expression of β-secretase, resulting in the formation of amyloid fibrils, which consequently aggregate into amyloid plaques (Cole and Vassar, 2007). Similarly, Aβ oligomers can in turn stimulate NF-κB activation in neurons and glial cells, forming a vicious cycle (Snow and Albensi, 2016).

Therapeutic strategies targeting NMDAR dysfunction in AD have evolved from simple receptor blockade to more nuanced approaches aimed at restoring physiological receptor function. Memantine remains the only FDA-approved drug targeting NMDARs for AD treatment, however its efficacy remains modest (Xia et al., 2010; Glasgow et al., 2017; Reisberg et al., 2003). Current research mainly focuses on subunit-selective modulators and spatially-targeted agents like NitroMemantine, a selective extrasynaptic NMDAR antagonist developed on the basis of memantine, which targets specific localized NMDARs, is of great potential for AD treatment.

6 NMDA receptor as a target for treating neurodegenerative diseases

NMDARs represent pivotal therapeutic targets for neurodegenerative diseases, governing excitotoxicity, synaptic dysfunction, and neuronal survival. Their dualistic nature: physiological activation supporting cognition versus pathological overstimulation driving degeneration, demands precisely calibrated interventions. Current therapeutic strategies prioritize activity-dependent modulation over complete receptor blockade. Subunit-selective agents now dominate therapeutic innovation: GluN2B-selective antagonists (e.g., ifenprodil and its derivatives) preferentially target neurodegenerative extrasynaptic receptors without impairing cognition in AD and HD models (Ugale et al., 2024), while GluN2A-positive allosteric modulators (PAMs) counteract synaptic depletion in late-stage disease (Yukawa et al., 2023).

Novel therapeutic strategies further expand the landscape: (1) Targeting kinases/phosphatases upstream of NMDARs (such as PKC activator bryostatin-1) or effectors downstream (such as DAPK1/NR2B uncoupler) (Zhang et al., 2020). Bryostatin-1 demonstrates efficacy in AD trials by reducing Aβ, promoting synaptogenesis, and suppressing oxidative stress, with favorable safety profiles enabling clinical application (Hongpaisan et al., 2011; Tian et al., 2023). In contrast, administration of a peptide NR2BCT1292–1,304 to uncouple the activated DAPK1 from the NMDA receptor complex protects against brain damage, which indicates that targeting DAPK1-NMDA receptor interaction can be considered as a practical strategy (Wang et al., 2017); (2) Receptor complex dissociation: Small molecules like ZL006 (uncoupling NMDAR/PSD-95) selectively block neurotoxic NO signaling without impairing physiological receptor function (Tao et al., 2020); (3) Neural circuit rebalancing (Ghatak et al., 2021), Restoring excitatory/inhibitory (E/I) imbalances caused by extrasynaptic NMDAR hyperactivity in autism and AD (Vico Varela et al., 2019; Schuch et al., 2016).

Despite robust preclinical evidence for NMDAR modulation in neurodegeneration, clinical translation has been hampered by intersecting pharmacological and biological barriers. Blood–brain barrier (BBB) penetration remains a primary bottleneck (Egunlusi and Joubert, 2024). Species divergence in receptor biology further complicates the clinical translation, rendering compounds optimized for murine receptors ineffective (such as the different effect of memantine in ALS on animal model and patients mentioned before). Crucially, achieving subunit-or localization-specific drug delivery in human patients is also a key barrier underlying the current NMDAR-based therapeutics. Emerging therapies aiming to overcome these hurdles include novel drug delivery systems such as lipid nanoparticles, loaded with riluzole (Bondì et al., 2010) and dopamine (Ortega Martínez et al., 2024), have shown promising results in increasing drug bioavailability in the CNS for ALS and PD treatment.

The future of NMDAR-targeted therapy lies in personalized combinatorial approaches: integration of subunit-selective drugs, neuroprotective agents, and disease-modifying treatments tailored to disease stage and specific pathology of each neurodegenerative disorder. By addressing both the excitotoxic mechanisms and the broader cellular context of NMDAR dysfunction, these strategies hold significant potential to slow or even halt neurodegeneration.

7 Discussion

The multifaceted role of NMDA receptors in neurodegenerative diseases has emerged as a central paradigm in understanding both the pathogenesis and potential treatment strategies for these neurodegenerative diseases. Our synthesis of current evidence reveals that NMDAR dysfunction operates through a complex, interconnected network of mechanisms that vary across different neurodegenerative disorders while sharing common pathological themes. At the core of this dysregulation lies the delicate balance between synaptic and extrasynaptic NMDAR signaling, a delicate balance that becomes profoundly disrupted in disease states. The consequences of this imbalance manifest through multiple converging pathways: excitotoxic calcium overload, oxidative stress, mitochondrial dysfunction, impaired synaptic plasticity, and maladaptive transcriptional changes.

Though the application of memantine and other NMDAR antagonists in different neurodegenerative diseases has shown certain potential, however, the efficacy of existing NMDAR antagonists is still limited and often result in significant side effects, such as euphoria, psychotic symptoms and increased blood pressure, indicating gaps in our current understanding of the diseases and the complexity of NMDAR functions (Muir, 2006). Recent advances in structural biology and receptor pharmacology have enabled the design of compounds with unprecedented specificity for particular NMDAR subtypes and locations. GluN2B-selective antagonists such as ifenprodil and its deriavtives (Gogas, 2006) represent a significant step forward by preferentially targeting receptors implicated in neurodegeneration while sparing those essential for cognitive function. Several recent studies have also investigated the potential of novel GluN2A-targeting positive allosteric modulators such as AGE-718 and 6-methylpyridin-2-one (Yukawa et al., 2023; Beckley et al., 2024), which also offers a complementary strategy to bolster synaptic resilience. Together, these approaches aim to restore the physiological balance between neuroprotective and neurotoxic NMDAR signaling.

Beyond direct receptor modulation, innovative strategies targeting downstream effectors (such as DAPK1) or upstream kinases/phosphatases (such as PKC) offer complementary value. Small molecules that disrupt the GluN2B-PSD95-nNOS complex such as ZL006 demonstrate how specific protein–protein interactions can be targeted to block neurotoxic signaling while preserving physiological receptor function.

The future of NMDAR-targeted therapy lies in precision medicine frameworks, considering the patient-specific genetic and molecular profiles to tailor NMDAR-targeting therapies. Another major frontier is the optimization of drug delivery to overcome the blood–brain barrier while maintaining therapeutic concentrations in relevant brain regions. The emergence of novel delivery systems, such as nanoparticle carriers holds promise for addressing this challenge. Perhaps most importantly, future therapies must account for the dynamic nature of NMDAR changes throughout disease progression. The receptor alterations that drive early synaptic dysfunction may differ substantially from those mediating late-stage neuronal death, suggesting that optimal interventions may need to evolve with disease progression.

The remarkable progress in understanding NMDAR biology over the past decades has transformed our approach to understand and treat neurodegenerative diseases. From viewing these receptors primarily as mediators of excitotoxicity, we now appreciate their roles in diverse pathological processes ranging from protein misfolding to neuroinflammation. This expanded understanding has given rise to a new generation of therapeutic strategies that seek not just to block excessive NMDAR activity, but to restore the delicate balance of synaptic and extrasynaptic signaling. As we continue to unravel the complexities of NMDAR regulation in health and disease, the prospect of developing truly disease-modifying treatments grows increasingly tangible. The path forward will require continued collaboration across disciplines, from structural biology to clinical neurology. By building on the foundations laid by current research and embracing the challenges that remain, we may finally be able to translate our knowledge of NMDAR mechanisms into transformative therapies for neurodegenerative diseases.

Author contributions

KZ: Formal analysis, Investigation, Methodology, Visualization, Writing – original draft. MW: Writing – original draft. XN: Writing – original draft. SZ: Writing – original draft. HL: Conceptualization, Funding acquisition, Visualization, Writing – review & editing. YA: Writing – review & editing. HZ: Funding acquisition, Writing – review & editing.

Funding

The author(s) declare that financial support was received for the research and/or publication of this article. This work was supported by grants from the National Natural Science Foundation of China (Grants: 32200795 to HL and 81801368 to HZ); Natural Science Foundation of Hubei Province (2022CFB608 to HL and 2024AFB593 to HZ).

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Generative AI statement

The authors declare that no Gen AI was used in the creation of this manuscript.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

Aksenov, M., Aksenova, M. V., Harris, M. E., Hensley, K., Butterfield, D. A., and Carney, J. M. (1995). Enhancement of beta-amyloid peptide a beta(1-40)-mediated neurotoxicity by glutamine synthetase. J. Neurochem. 65, 1899–1902.

Google Scholar

Alleva, C., Kovalev, K., Astashkin, R., Berndt, M. I., Baeken, C., Balandin, T., et al. (2020). Na(+)-dependent gate dynamics and electrostatic attraction ensure substrate coupling in glutamate transporters. Sci. Adv. 6:9854. doi: 10.1126/sciadv.aba9854

PubMed Abstract | Crossref Full Text | Google Scholar

Alleva, C., Machtens, J. P., Kortzak, D., Weyand, I., and Fahlke, C. (2022). Molecular basis of coupled transport and anion conduction in excitatory amino acid transporters. Neurochem. Res. 47, 9–22. doi: 10.1007/s11064-021-03252-x

PubMed Abstract | Crossref Full Text | Google Scholar

Amidfar, M., de Oliveira, J., Kucharska, E., Budni, J., and Kim, Y. K. (2020). The role of CREB and BDNF in neurobiology and treatment of Alzheimer's disease. Life Sci. 257:118020. doi: 10.1016/j.lfs.2020.118020

PubMed Abstract | Crossref Full Text | Google Scholar

Andersen, J. V., Markussen, K. H., Jakobsen, E., Schousboe, A., Waagepetersen, H. S., Rosenberg, P. A., et al. (2021). Glutamate metabolism and recycling at the excitatory synapse in health and neurodegeneration. Neuropharmacology 196:108719. doi: 10.1016/j.neuropharm.2021.108719

Crossref Full Text | Google Scholar

Babaei, P. (2021). NMDA and AMPA receptors dysregulation in Alzheimer's disease. Eur. J. Pharmacol. 908:174310. doi: 10.1016/j.ejphar.2021.174310

PubMed Abstract | Crossref Full Text | Google Scholar

Bading, H. (2017). Therapeutic targeting of the pathological triad of extrasynaptic NMDA receptor signaling in neurodegenerations. J. Exp. Med. 214, 569–578. doi: 10.1084/jem.20161673

PubMed Abstract | Crossref Full Text | Google Scholar

Bannerman, D. M., Sprengel, R., Sanderson, D. J., McHugh, S. B., Rawlins, J. N., Monyer, H., et al. (2014). Hippocampal synaptic plasticity, spatial memory and anxiety. Nat. Rev. Neurosci. 15, 181–192. doi: 10.1038/nrn3677

PubMed Abstract | Crossref Full Text | Google Scholar

Bartolotti, N., Segura, L., and Lazarov, O. (2016). Diminished CRE-induced plasticity is linked to memory deficits in familial Alzheimer's disease mice. J. Alzheimers Dis. 50, 477–489. doi: 10.3233/JAD-150650

PubMed Abstract | Crossref Full Text | Google Scholar

Beal, M. F. (1995). Aging, energy, and oxidative stress in neurodegenerative diseases. Ann. Neurol. 38, 357–366. doi: 10.1002/ana.410380304

PubMed Abstract | Crossref Full Text | Google Scholar

Beal, M. F. (1998). Excitotoxicity and nitric oxide in Parkinson's disease pathogenesis. Ann. Neurol. 44, S110–S114. doi: 10.1002/ana.410440716

PubMed Abstract | Crossref Full Text | Google Scholar

Beckley, J. T., Aman, T. K., Ackley, M. A., Kazdoba, T. M., Lewis, M. C., Smith, A. C., et al. (2024). Pharmacological characterization of SAGE-718, a novel positive allosteric modulator of N-methyl-d-aspartate receptors. Br. J. Pharmacol. 181, 1028–1050. doi: 10.1111/bph.16235

PubMed Abstract | Crossref Full Text | Google Scholar

Bhai, S., Levine, T., Moore, D., Bowser, R., Heim, A. J., Walsh, M., et al. (2025). A 40-week phase 2B randomized, multicenter, double-blind, placebo-controlled study evaluating the safety and efficacy of memantine in amyotrophic lateral sclerosis. Muscle Nerve 71, 63–72. doi: 10.1002/mus.28287

PubMed Abstract | Crossref Full Text | Google Scholar

Bliss, T. V., and Collingridge, G. L. (1993). A synaptic model of memory: long-term potentiation in the hippocampus. Nature 361, 31–39. doi: 10.1038/361031a0

PubMed Abstract | Crossref Full Text | Google Scholar

Bondì, M. L., Craparo, E. F., Giammona, G., and Drago, F. (2010). Brain-targeted solid lipid nanoparticles containing riluzole: preparation, characterization and biodistribution. Nanomedicine (Lond.) 5, 25–32. doi: 10.2217/nnm.09.67

PubMed Abstract | Crossref Full Text | Google Scholar

Brown, G. C., and Bal-Price, A. (2003). Inflammatory neurodegeneration mediated by nitric oxide, glutamate, and mitochondria. Mol. Neurobiol. 27, 325–355. doi: 10.1385/MN:27:3:325

PubMed Abstract | Crossref Full Text | Google Scholar

Burk, K., and Pasterkamp, R. J. (2019). Disrupted neuronal trafficking in amyotrophic lateral sclerosis. Acta Neuropathol. 137, 859–877. doi: 10.1007/s00401-019-01964-7

PubMed Abstract | Crossref Full Text | Google Scholar

Catania, M. V., Aronica, E., Yankaya, B., and Troost, D. (2001). Increased expression of neuronal nitric oxide synthase spliced variants in reactive astrocytes of amyotrophic lateral sclerosis human spinal cord. J. Neurosci. 21:RC148. doi: 10.1523/JNEUROSCI.21-11-j0002.2001

PubMed Abstract | Crossref Full Text | Google Scholar

Cenini, G., Lloret, A., and Cascella, R. (2019). Oxidative stress in neurodegenerative diseases: from a mitochondrial point of view. Oxidative Med. Cell. Longev. 2019, 1–18. doi: 10.1155/2019/2105607

PubMed Abstract | Crossref Full Text | Google Scholar

Cepeda, C., Wu, N., Andre, V. M., Cummings, D. M., and Levine, M. S. (2007). The corticostriatal pathway in Huntington's disease. Prog. Neurobiol. 81, 253–271. doi: 10.1016/j.pneurobio.2006.11.001

PubMed Abstract | Crossref Full Text | Google Scholar

Chan, S. L., and Mattson, M. P. (1999). Caspase and calpain substrates: roles in synaptic plasticity and cell death. J. Neurosci. Res. 58, 167–190. doi: 10.1002/(SICI)1097-4547(19991001)58:1<>3.0.CO;2-K

PubMed Abstract | Crossref Full Text | Google Scholar

Chatterton, J. E., Awobuluyi, M., Premkumar, L. S., Takahashi, H., Talantova, M., Shin, Y., et al. (2002). Excitatory glycine receptors containing the NR3 family of NMDA receptor subunits. Nature 415, 793–798. doi: 10.1038/nature715

PubMed Abstract | Crossref Full Text | Google Scholar

Chen, X., Levy, J. M., Hou, A., Winters, C., Azzam, R., Sousa, A. A., et al. (2015). PSD-95 family MAGUKs are essential for anchoring AMPA and NMDA receptor complexes at the postsynaptic density. Proc. Natl. Acad. Sci. USA 112, E6983–E6992. doi: 10.1073/pnas.1517045112

PubMed Abstract | Crossref Full Text | Google Scholar

Chou, T. H., Epstein, M., Fritzemeier, R. G., Akins, N. S., Paladugu, S., Ullman, E. Z., et al. (2024). Molecular mechanism of ligand gating and opening of NMDA receptor. Nature 632, 209–217. doi: 10.1038/s41586-024-07742-0

PubMed Abstract | Crossref Full Text | Google Scholar

Cole, S. L., and Vassar, R. (2007). The Alzheimer's disease beta-secretase enzyme, BACE1. Mol. Neurodegener. 2:22. doi: 10.1186/1750-1326-2-22

PubMed Abstract | Crossref Full Text | Google Scholar

Crawley, O., Conde-Dusman, M. J., and Pérez-Otaño, I. (2022). GluN3A NMDA receptor subunits: more enigmatic than ever? J. Physiol. 600, 261–276. doi: 10.1113/JP280879

PubMed Abstract | Crossref Full Text | Google Scholar

Crosio, C., Valle, C., Casciati, A., Iaccarino, C., and Carrì, M. T. (2011). Astroglial inhibition of NF-κB does not ameliorate disease onset and progression in a mouse model for amyotrophic lateral sclerosis (ALS). PLoS One 6:e17187. doi: 10.1371/journal.pone.0017187

PubMed Abstract | Crossref Full Text | Google Scholar

Cui, L., Jeong, H., Borovecki, F., Parkhurst, C. N., Tanese, N., and Krainc, D. (2006). Transcriptional repression of PGC-1alpha by mutant huntingtin leads to mitochondrial dysfunction and neurodegeneration. Cell 127, 59–69. doi: 10.1016/j.cell.2006.09.015

PubMed Abstract | Crossref Full Text | Google Scholar

Cull-Candy, S., Brickley, S., and Farrant, M. (2001). NMDA receptor subunits: diversity, development and disease. Curr. Opin. Neurobiol. 11, 327–335. doi: 10.1016/S0959-4388(00)00215-4

PubMed Abstract | Crossref Full Text | Google Scholar

Dawson, V. L., and Dawson, T. M. (1996). Nitric oxide neurotoxicity. J. Chem. Neuroanat. 10, 179–190. doi: 10.1016/0891-0618(96)00148-2

PubMed Abstract | Crossref Full Text | Google Scholar

Dawson, V. L., Dawson, T. M., London, E. D., Bredt, D. S., and Snyder, S. H. (1991). Nitric oxide mediates glutamate neurotoxicity in primary cortical cultures. Proc. Natl. Acad. Sci. USA 88, 6368–6371. doi: 10.1073/pnas.88.14.6368

PubMed Abstract | Crossref Full Text | Google Scholar

de Carvalho, M., Pinto, S., Costa, J., Evangelista, T., Ohana, B., and Pinto, A. (2010). A randomized, placebo-controlled trial of memantine for functional disability in amyotrophic lateral sclerosis. Amyotroph. Lateral Scler. 11, 456–460. doi: 10.3109/17482968.2010.498521

PubMed Abstract | Crossref Full Text | Google Scholar

De Felice, F. G., Velasco, P. T., Lambert, M. P., Viola, K., Fernandez, S. J., Ferreira, S. T., et al. (2007). Abeta oligomers induce neuronal oxidative stress through an N-methyl-D-aspartate receptor-dependent mechanism that is blocked by the Alzheimer drug memantine. J. Biol. Chem. 282, 11590–11601. doi: 10.1074/jbc.M607483200

PubMed Abstract | Crossref Full Text | Google Scholar

de Oca, M., and Balderas, P. (2018). Flux-independent NMDAR signaling: molecular mediators, cellular functions, and complexities. Int. J. Mol. Sci. 19:800. doi: 10.3390/ijms19123800

PubMed Abstract | Crossref Full Text | Google Scholar

Dembitsky, V. M., Gloriozova, T. A., and Poroikov, V. V. (2020). Pharmacological profile of natural and synthetic compounds with rigid adamantane-based scaffolds as potential agents for the treatment of neurodegenerative diseases. Biochem. Biophys. Res. Commun. 529, 1225–1241. doi: 10.1016/j.bbrc.2020.06.123

PubMed Abstract | Crossref Full Text | Google Scholar

Dong, X. X., Wang, Y., and Qin, Z. H. (2009). Molecular mechanisms of excitotoxicity and their relevance to pathogenesis of neurodegenerative diseases. Acta Pharmacol. Sin. 30, 379–387. doi: 10.1038/aps.2009.24

PubMed Abstract | Crossref Full Text | Google Scholar

Dong, B., Yue, Y., Dong, H., and Wang, Y. (2023). N-methyl-D-aspartate receptor hypofunction as a potential contributor to the progression and manifestation of many neurological disorders. Front. Mol. Neurosci. 16:1174738. doi: 10.3389/fnmol.2023.1174738

PubMed Abstract | Crossref Full Text | Google Scholar

Dunah, A. W., and Standaert, D. G. (2001). Dopamine D1 receptor-dependent trafficking of striatal NMDA glutamate receptors to the postsynaptic membrane. J. Neurosci. 21, 5546–5558. doi: 10.1523/JNEUROSCI.21-15-05546.2001

PubMed Abstract | Crossref Full Text | Google Scholar

Dunah, A. W., Wang, Y., Yasuda, R. P., Kameyama, K., Huganir, R. L., Wolfe, B. B., et al. (2000). Alterations in subunit expression, composition, and phosphorylation of striatal N-methyl-D-aspartate glutamate receptors in a rat 6-hydroxydopamine model of Parkinson's disease. Mol. Pharmacol. 57, 342–352. doi: 10.1016/S0026-895X(24)23206-5

PubMed Abstract | Crossref Full Text | Google Scholar

Duncan, A. J., and Heales, S. J. (2005). Nitric oxide and neurological disorders. Mol. Asp. Med. 26, 67–96. doi: 10.1016/j.mam.2004.09.004

PubMed Abstract | Crossref Full Text | Google Scholar

Durante, V., de Iure, A., Loffredo, V., Vaikath, N., De Risi, M., Paciotti, S., et al. (2019). Alpha-synuclein targets GluN2A NMDA receptor subunit causing striatal synaptic dysfunction and visuospatial memory alteration. Brain 142, 1365–1385. doi: 10.1093/brain/awz065

PubMed Abstract | Crossref Full Text | Google Scholar

Egunlusi, A. O., and Joubert, J. (2024). NMDA receptor antagonists: emerging insights into molecular mechanisms and clinical applications in neurological disorders. Pharmaceuticals (Basel) 17:639. doi: 10.3390/ph17050639

PubMed Abstract | Crossref Full Text | Google Scholar

Escamilla, S., Badillos, R., Comella, J. X., Solé, M., Pérez-Otaño, I., Mut, J. V. S., et al. (2024). Synaptic and extrasynaptic distribution of NMDA receptors in the cortex of Alzheimer's disease patients. Alzheimers Dement. 20, 8231–8245. doi: 10.1002/alz.14125

PubMed Abstract | Crossref Full Text | Google Scholar

Esposito, Z., Belli, L., Toniolo, S., Sancesario, G., Bianconi, C., and Martorana, A. (2013). Amyloid β, glutamate, excitotoxicity in Alzheimer's disease: are we on the right track? CNS Neurosci. Ther. 19, 549–555. doi: 10.1111/cns.12095

PubMed Abstract | Crossref Full Text | Google Scholar

Fan, J., Cowan, C. M., Zhang, L. Y., Hayden, M. R., and Raymond, L. A. (2009). Interaction of postsynaptic density protein-95 with NMDA receptors influences excitotoxicity in the yeast artificial chromosome mouse model of Huntington's disease. J. Neurosci. 29, 10928–10938. doi: 10.1523/JNEUROSCI.2491-09.2009

PubMed Abstract | Crossref Full Text | Google Scholar

Fan, M. M., and Raymond, L. A. (2007). N-methyl-D-aspartate (NMDA) receptor function and excitotoxicity in Huntington's disease. Prog. Neurobiol. 81, 272–293. doi: 10.1016/j.pneurobio.2006.11.003

PubMed Abstract | Crossref Full Text | Google Scholar

Feldman, E. L., Goutman, S. A., Petri, S., Mazzini, L., Savelieff, M. G., Shaw, P. J., et al. (2022). Amyotrophic lateral sclerosis. Lancet 400, 1363–1380. doi: 10.1016/S0140-6736(22)01272-7

PubMed Abstract | Crossref Full Text | Google Scholar

Fernandes, H. B., and Raymond, L. A.. Frontiers in neuroscience NMDA Receptors and Huntington’s Disease. In: A. M. DongenVan, editor. Biology of the NMDA receptor, Boca Raton (FL): CRC press/Taylor & Francis. (2009).

Google Scholar

Frakes, A. E., Ferraiuolo, L., Haidet-Phillips, A. M., Schmelzer, L., Braun, L., Miranda, C. J., et al. (2014). Microglia induce motor neuron death via the classical NF-κB pathway in amyotrophic lateral sclerosis. Neuron 81, 1009–1023. doi: 10.1016/j.neuron.2014.01.013

PubMed Abstract | Crossref Full Text | Google Scholar

Garzon, D. J., and Fahnestock, M. (2007). Oligomeric amyloid decreases basal levels of brain-derived neurotrophic factor (BDNF) mRNA via specific downregulation of BDNF transcripts IV and V in differentiated human neuroblastoma cells. J. Neurosci. 27, 2628–2635. doi: 10.1523/JNEUROSCI.5053-06.2007

PubMed Abstract | Crossref Full Text | Google Scholar

Gaschler, M. M., and Stockwell, B. R. (2017). Lipid peroxidation in cell death. Biochem. Biophys. Res. Commun. 482, 419–425. doi: 10.1016/j.bbrc.2016.10.086

PubMed Abstract | Crossref Full Text | Google Scholar

Ghasemi, M., Mayasi, Y., Hannoun, A., Eslami, S. M., and Carandang, R. (2018). Nitric oxide and mitochondrial function in neurological diseases. Neuroscience 376, 48–71. doi: 10.1016/j.neuroscience.2018.02.017

PubMed Abstract | Crossref Full Text | Google Scholar

Ghatak, S., Talantova, M., McKercher, S. R., and Lipton, S. A. (2021). Novel therapeutic approach for excitatory/inhibitory imbalance in neurodevelopmental and neurodegenerative diseases. Annu. Rev. Pharmacol. Toxicol. 61, 701–721. doi: 10.1146/annurev-pharmtox-032320-015420

PubMed Abstract | Crossref Full Text | Google Scholar

Gladding, C. M., and Raymond, L. A. (2011). Mechanisms underlying NMDA receptor synaptic/extrasynaptic distribution and function. Mol. Cell. Neurosci. 48, 308–320. doi: 10.1016/j.mcn.2011.05.001

PubMed Abstract | Crossref Full Text | Google Scholar

Glasgow, N. G., Povysheva, N. V., Azofeifa, A. M., and Johnson, J. W. (2017). Memantine and ketamine differentially Alter NMDA receptor desensitization. J. Neurosci. 37, 9686–9704. doi: 10.1523/JNEUROSCI.1173-17.2017

PubMed Abstract | Crossref Full Text | Google Scholar

Gogas, K. R. (2006). Glutamate-based therapeutic approaches: NR2B receptor antagonists. Curr. Opin. Pharmacol. 6, 68–74. doi: 10.1016/j.coph.2005.11.001

PubMed Abstract | Crossref Full Text | Google Scholar

Greenamyre, J. T., and O'Brien, C. F. (1991). N-methyl-D-aspartate antagonists in the treatment of Parkinson's disease. Arch. Neurol. 48, 977–981. doi: 10.1001/archneur.1991.00530210109030

PubMed Abstract | Crossref Full Text | Google Scholar

Greer, P. L., and Greenberg, M. E. (2008). From synapse to nucleus: calcium-dependent gene transcription in the control of synapse development and function. Neuron 59, 846–860. doi: 10.1016/j.neuron.2008.09.002

PubMed Abstract | Crossref Full Text | Google Scholar

Groc, L., Bard, L., and Choquet, D. (2009). Surface trafficking of N-methyl-D-aspartate receptors: physiological and pathological perspectives. Neuroscience 158, 4–18. doi: 10.1016/j.neuroscience.2008.05.029

PubMed Abstract | Crossref Full Text | Google Scholar

Hansen, K. B., Yi, F., Perszyk, R. E., Furukawa, H., Wollmuth, L. P., Gibb, A. J., et al. (2018). Structure, function, and allosteric modulation of NMDA receptors. J. Gen. Physiol. 150, 1081–1105. doi: 10.1085/jgp.201812032

PubMed Abstract | Crossref Full Text | Google Scholar

Hanson, J. E., Ma, K., Elstrott, J., Weber, M., Saillet, S., Khan, A. S., et al. (2020). GluN2A NMDA receptor enhancement improves brain oscillations, synchrony, and cognitive functions in Dravet syndrome and Alzheimer's disease models. Cell Rep. 30, 381–96.e4. doi: 10.1016/j.celrep.2019.12.030

PubMed Abstract | Crossref Full Text | Google Scholar

Hardiman, O., Al-Chalabi, A., Chio, A., Corr, E. M., Logroscino, G., Robberecht, W., et al. (2017). Amyotrophic lateral sclerosis. Nat. Rev. Dis. Primers 3:17071. doi: 10.1038/nrdp.2017.71

PubMed Abstract | Crossref Full Text | Google Scholar

Hardingham, G. E., and Bading, H. (2003). The yin and Yang of NMDA receptor signalling. Trends Neurosci. 26, 81–89. doi: 10.1016/S0166-2236(02)00040-1

PubMed Abstract | Crossref Full Text | Google Scholar

Hardingham, G. E., and Bading, H. (2010). Synaptic versus extrasynaptic NMDA receptor signalling: implications for neurodegenerative disorders. Nat. Rev. Neurosci. 11, 682–696. doi: 10.1038/nrn2911

PubMed Abstract | Crossref Full Text | Google Scholar

Hardingham, G. E., Fukunaga, Y., and Bading, H. (2002). Extrasynaptic NMDARs oppose synaptic NMDARs by triggering CREB shut-off and cell death pathways. Nat. Neurosci. 5, 405–414. doi: 10.1038/nn835

PubMed Abstract | Crossref Full Text | Google Scholar

Hayes, M. T. (2019). Parkinson's disease and parkinsonism. Am. J. Med. 132, 802–807. doi: 10.1016/j.amjmed.2019.03.001

PubMed Abstract | Crossref Full Text | Google Scholar

Heath, P. R., and Shaw, P. J. (2002). Update on the glutamatergic neurotransmitter system and the role of excitotoxicity in amyotrophic lateral sclerosis. Muscle Nerve 26, 438–458. doi: 10.1002/mus.10186

PubMed Abstract | Crossref Full Text | Google Scholar

Henson, M. A., Roberts, A. C., Pérez-Otaño, I., and Philpot, B. D. (2010). Influence of the NR3A subunit on NMDA receptor functions. Prog. Neurobiol. 91, 23–37. doi: 10.1016/j.pneurobio.2010.01.004

PubMed Abstract | Crossref Full Text | Google Scholar

Hongpaisan, J., Sun, M. K., and Alkon, D. L. (2011). PKC epsilon activation prevents synaptic loss, Abeta elevation, and cognitive deficits in Alzheimer's disease transgenic mice. J. Neurosci. 31, 630–643. doi: 10.1523/JNEUROSCI.5209-10.2011

Crossref Full Text | Google Scholar

Hoover, B. R., Reed, M. N., Su, J., Penrod, R. D., Kotilinek, L. A., Grant, M. K., et al. (2010). Tau mislocalization to dendritic spines mediates synaptic dysfunction independently of neurodegeneration. Neuron 68, 1067–1081. doi: 10.1016/j.neuron.2010.11.030

PubMed Abstract | Crossref Full Text | Google Scholar

Howland, D. S., Liu, J., She, Y., Goad, B., Maragakis, N. J., Kim, B., et al. (2002). Focal loss of the glutamate transporter EAAT2 in a transgenic rat model of SOD1 mutant-mediated amyotrophic lateral sclerosis (ALS). Proc. Natl. Acad. Sci. USA 99, 1604–1609. doi: 10.1073/pnas.032539299

PubMed Abstract | Crossref Full Text | Google Scholar

Islam, M. T. (2017). Oxidative stress and mitochondrial dysfunction-linked neurodegenerative disorders. Neurol. Res. 39, 73–82. doi: 10.1080/01616412.2016.1251711

PubMed Abstract | Crossref Full Text | Google Scholar

Jiang, J., Wang, Y., and Deng, M. (2022). New developments and opportunities in drugs being trialed for amyotrophic lateral sclerosis from 2020 to 2022. Front. Pharmacol. 13:1054006. doi: 10.3389/fphar.2022.1054006

PubMed Abstract | Crossref Full Text | Google Scholar

Johnson, J. W., and Kotermanski, S. E. (2006). Mechanism of action of memantine. Curr. Opin. Pharmacol. 6, 61–67. doi: 10.1016/j.coph.2005.09.007

PubMed Abstract | Crossref Full Text | Google Scholar

Jones, S., and Gibb, A. J. (2005). Functional NR2B- and NR2D-containing NMDA receptor channels in rat substantia nigra dopaminergic neurones. J. Physiol. 569, 209–221. doi: 10.1113/jphysiol.2005.095554

PubMed Abstract | Crossref Full Text | Google Scholar

Joo, I. S., Hwang, D. H., Seok, J. I., Shin, S. K., and Kim, S. U. (2007). Oral administration of memantine prolongs survival in a transgenic mouse model of amyotrophic lateral sclerosis. J. Clin. Neurol. 3, 181–186. doi: 10.3988/jcn.2007.3.4.181

PubMed Abstract | Crossref Full Text | Google Scholar

Kalia, L. V., and Lang, A. E. (2015). Parkinson's disease. Lancet 386, 896–912. doi: 10.1016/S0140-6736(14)61393-3

PubMed Abstract | Crossref Full Text | Google Scholar

Källstig, E., McCabe, B. D., and Schneider, B. L. (2021). The links between ALS and NF-κB. Int. J. Mol. Sci. 22:875. doi: 10.3390/ijms22083875

PubMed Abstract | Crossref Full Text | Google Scholar

Kessels, H. W., Nabavi, S., and Malinow, R. (2013). Metabotropic NMDA receptor function is required for β-amyloid-induced synaptic depression. Proc. Natl. Acad. Sci. USA 110, 4033–4038. doi: 10.1073/pnas.1219605110

PubMed Abstract | Crossref Full Text | Google Scholar

Kim, A., Lalonde, K., Truesdell, A., Gomes Welter, P., Brocardo, P. S., Rosenstock, T. R., et al. (2021). New avenues for the treatment of Huntington's disease. Int. J. Mol. Sci. 22:363. doi: 10.3390/ijms22168363

PubMed Abstract | Crossref Full Text | Google Scholar

Kim, K., Lee, S. G., Kegelman, T. P., Su, Z. Z., Das, S. K., Dash, R., et al. (2011). Role of excitatory amino acid transporter-2 (EAAT2) and glutamate in neurodegeneration: opportunities for developing novel therapeutics. J. Cell. Physiol. 226, 2484–2493. doi: 10.1002/jcp.22609

PubMed Abstract | Crossref Full Text | Google Scholar

Kodis, E. J., Choi, S., Swanson, E., Ferreira, G., and Bloom, G. S. (2018). N-methyl-D-aspartate receptor-mediated calcium influx connects amyloid-β oligomers to ectopic neuronal cell cycle reentry in Alzheimer's disease. Alzheimers Dement. 14, 1302–1312. doi: 10.1016/j.jalz.2018.05.017

PubMed Abstract | Crossref Full Text | Google Scholar

Köhr, G. (2006). NMDA receptor function: subunit composition versus spatial distribution. Cell Tissue Res. 326, 439–446. doi: 10.1007/s00441-006-0273-6

PubMed Abstract | Crossref Full Text | Google Scholar

Kondori, N. R., Paul, P., Robbins, J. P., Liu, K., Hildyard, J. C. W., Wells, D. J., et al. (2018). Focus on the role of D-serine and D-amino acid oxidase in amyotrophic lateral sclerosis/motor neuron disease (ALS). Front. Mol. Biosci. 5:8. doi: 10.3389/fmolb.2018.00008

PubMed Abstract | Crossref Full Text | Google Scholar

Kulijewicz-Nawrot, M., Syková, E., Chvátal, A., Verkhratsky, A., and Rodríguez, J. J. (2013). Astrocytes and glutamate homoeostasis in Alzheimer's disease: a decrease in glutamine synthetase, but not in glutamate transporter-1, in the prefrontal cortex. ASN Neuro 5, 273–282. doi: 10.1042/AN20130017

PubMed Abstract | Crossref Full Text | Google Scholar

Landwehrmeyer, G. B., Standaert, D. G., Testa, C. M., Penney, J. B. Jr., and Young, A. B. (1995). NMDA receptor subunit mRNA expression by projection neurons and interneurons in rat striatum. J. Neurosci. 15, 5297–5307. doi: 10.1523/JNEUROSCI.15-07-05297.1995

PubMed Abstract | Crossref Full Text | Google Scholar

Lane, C. A., Hardy, J., and Schott, J. M. (2018). Alzheimer's disease. Eur. J. Neurol. 25, 59–70. doi: 10.1111/ene.13439

PubMed Abstract | Crossref Full Text | Google Scholar

Lau, A., and Tymianski, M. (2010). Glutamate receptors, neurotoxicity and neurodegeneration. Pflugers Arch. 460, 525–542. doi: 10.1007/s00424-010-0809-1

PubMed Abstract | Crossref Full Text | Google Scholar

Li, L., Fan, M., Icton, C. D., Chen, N., Leavitt, B. R., Hayden, M. R., et al. (2003). Role of NR2B-type NMDA receptors in selective neurodegeneration in Huntington disease. Neurobiol. Aging 24, 1113–1121. doi: 10.1016/j.neurobiolaging.2003.04.003

PubMed Abstract | Crossref Full Text | Google Scholar

Li, D., Specht, C. G., Waites, C. L., Butler-Munro, C., Leal-Ortiz, S., Foote, J. W., et al. (2011). SAP97 directs NMDA receptor spine targeting and synaptic plasticity. J. Physiol. 589, 4491–4510. doi: 10.1113/jphysiol.2011.215566

PubMed Abstract | Crossref Full Text | Google Scholar

Liang, C. L., Sinton, C. M., Sonsalla, P. K., and German, D. C. (1996). Midbrain dopaminergic neurons in the mouse that contain calbindin-D28k exhibit reduced vulnerability to MPTP-induced neurodegeneration. Neurodegeneration 5, 313–318. doi: 10.1006/neur.1996.0042

PubMed Abstract | Crossref Full Text | Google Scholar

Lin, M. T., and Beal, M. F. (2006). Mitochondrial dysfunction and oxidative stress in neurodegenerative diseases. Nature 443, 787–795. doi: 10.1038/nature05292

PubMed Abstract | Crossref Full Text | Google Scholar

Lindsley, C. W., Shipe, W. D., Wolkenberg, S. E., Theberge, C. R., Williams, D. L. Jr., Sur, C., et al. (2006). Progress towards validating the NMDA receptor hypofunction hypothesis of schizophrenia. Curr. Top. Med. Chem. 6, 771–785. doi: 10.2174/156802606777057599

PubMed Abstract | Crossref Full Text | Google Scholar

Lohmann, C., and Kessels, H. W. (2014). The developmental stages of synaptic plasticity. J. Physiol. 592, 13–31. doi: 10.1113/jphysiol.2012.235119

PubMed Abstract | Crossref Full Text | Google Scholar

Lovell, M. A., Bradley, M. A., and Fister, S. X. (2012). 4-Hydroxyhexenal (HHE) impairs glutamate transport in astrocyte cultures. J. Alzheimers Dis. 32, 139–146. doi: 10.3233/JAD-2012-120409

PubMed Abstract | Crossref Full Text | Google Scholar

Lüscher, C., and Malenka, R. C. (2012). NMDA receptor-dependent long-term potentiation and long-term depression (LTP/LTD). Cold Spring Harb. Perspect. Biol. 4:710. doi: 10.1101/cshperspect.a005710

PubMed Abstract | Crossref Full Text | Google Scholar

Mahmoud, S., Gharagozloo, M., Simard, C., and Gris, D. (2019). Astrocytes maintain glutamate homeostasis in the CNS by controlling the balance between glutamate uptake and release. Cells 8:184. doi: 10.3390/cells8020184

PubMed Abstract | Crossref Full Text | Google Scholar

Malinski, T. (2007). Nitric oxide and nitroxidative stress in Alzheimer's disease. J. Alzheimers Dis. 11, 207–218. doi: 10.3233/JAD-2007-11208

PubMed Abstract | Crossref Full Text | Google Scholar

Marco, S., Giralt, A., Petrovic, M. M., Pouladi, M. A., Martínez-Turrillas, R., Martínez-Hernández, J., et al. (2013). Suppressing aberrant GluN3A expression rescues synaptic and behavioral impairments in Huntington's disease models. Nat. Med. 19, 1030–1038. doi: 10.1038/nm.3246

PubMed Abstract | Crossref Full Text | Google Scholar

McColgan, P., and Tabrizi, S. J. (2018). Huntington's disease: a clinical review. Eur. J. Neurol. 25, 24–34. doi: 10.1111/ene.13413

PubMed Abstract | Crossref Full Text | Google Scholar

Mellone, M., Stanic, J., Hernandez, L. F., Iglesias, E., Zianni, E., Longhi, A., et al. (2015). NMDA receptor GluN2A/GluN2B subunit ratio as synaptic trait of levodopa-induced dyskinesias: from experimental models to patients. Front. Cell. Neurosci. 9:245. doi: 10.3389/fncel.2015.00245

PubMed Abstract | Crossref Full Text | Google Scholar

Mellone, M., Zianni, E., Stanic, J., Campanelli, F., Marino, G., Ghiglieri, V., et al. (2019). NMDA receptor GluN2D subunit participates to levodopa-induced dyskinesia pathophysiology. Neurobiol. Dis. 121, 338–349. doi: 10.1016/j.nbd.2018.09.021

PubMed Abstract | Crossref Full Text | Google Scholar

Melzer, S., and Monyer, H. (2020). Diversity and function of corticopetal and corticofugal GABAergic projection neurons. Nat. Rev. Neurosci. 21, 499–515. doi: 10.1038/s41583-020-0344-9

PubMed Abstract | Crossref Full Text | Google Scholar

Menzies, F. M., Ince, P. G., and Shaw, P. J. (2002). Mitochondrial involvement in amyotrophic lateral sclerosis. Neurochem. Int. 40, 543–551. doi: 10.1016/s0197-0186(01)00125-5

PubMed Abstract | Crossref Full Text | Google Scholar

Merello, M., Nouzeilles, M. I., Cammarota, A., and Leiguarda, R. (1999). Effect of memantine (NMDA antagonist) on Parkinson's disease: a double-blind crossover randomized study. Clin. Neuropharmacol. 22, 273–276.

Google Scholar

Metman, L. V., Del Dotto, P., van den Munckhof, P., Fang, J., Mouradian, M. M., and Chase, T. N. (1998). Amantadine as treatment for dyskinesias and motor fluctuations in Parkinson's disease. Nervenheilkunde 17, 480–484.

Google Scholar

Mi, Z., Abrahamson, E. E., Ryu, A. Y., Malek-Ahmadi, M., Kofler, J. K., Fish, K. N., et al. (2023). Vesicular glutamate transporter changes in the cortical default mode network during the clinical and pathological progression of Alzheimer's disease. J. Alzheimers Dis. 94, 227–246. doi: 10.3233/JAD-221063

PubMed Abstract | Crossref Full Text | Google Scholar

Miller, R. G., Mitchell, J. D., and Moore, D. H. (2012). Riluzole for amyotrophic lateral sclerosis (ALS)/motor neuron disease (MND). Cochrane Database Syst. Rev. 3:Cd001447.

Google Scholar

Milnerwood, A. J., Gladding, C. M., Pouladi, M. A., Kaufman, A. M., Hines, R. M., Boyd, J. D., et al. (2010). Early increase in extrasynaptic NMDA receptor signaling and expression contributes to phenotype onset in Huntington's disease mice. Neuron 65, 178–190. doi: 10.1016/j.neuron.2010.01.008

PubMed Abstract | Crossref Full Text | Google Scholar

Mitchell, J., Paul, P., Chen, H. J., Morris, A., Payling, M., Falchi, M., et al. (2010). Familial amyotrophic lateral sclerosis is associated with a mutation in D-amino acid oxidase. Proc. Natl. Acad. Sci. USA 107, 7556–7561. doi: 10.1073/pnas.0914128107

PubMed Abstract | Crossref Full Text | Google Scholar

Monyer, H., Burnashev, N., Laurie, D. J., Sakmann, B., and Seeburg, P. H. (1994). Developmental and regional expression in the rat brain and functional properties of four NMDA receptors. Neuron 12, 529–540. doi: 10.1016/0896-6273(94)90210-0

PubMed Abstract | Crossref Full Text | Google Scholar

Morris, R. G. (2013). NMDA receptors and memory encoding. Neuropharmacology 74, 32–40. doi: 10.1016/j.neuropharm.2013.04.014

PubMed Abstract | Crossref Full Text | Google Scholar

Muir, K. W. (2006). Glutamate-based therapeutic approaches: clinical trials with NMDA antagonists. Curr. Opin. Pharmacol. 6, 53–60. doi: 10.1016/j.coph.2005.12.002

PubMed Abstract | Crossref Full Text | Google Scholar

Murphy, M. P. (2009). How mitochondria produce reactive oxygen species. Biochem. J. 417, 1–13. doi: 10.1042/BJ20081386

PubMed Abstract | Crossref Full Text | Google Scholar

Murphy-Royal, C., Dupuis, J., Groc, L., and Oliet, S. H. R. (2017). Astroglial glutamate transporters in the brain: regulating neurotransmitter homeostasis and synaptic transmission. J. Neurosci. Res. 95, 2140–2151. doi: 10.1002/jnr.24029

PubMed Abstract | Crossref Full Text | Google Scholar

Nabavi, S., Fox, R., Alfonso, S., Aow, J., and Malinow, R. (2014). GluA1 trafficking and metabotropic NMDA: addressing results from other laboratories inconsistent with ours. Philos. Trans. R. Soc. Lond. Ser. B Biol. Sci. 369:20130145. doi: 10.1098/rstb.2013.0145

PubMed Abstract | Crossref Full Text | Google Scholar

Nakazawa, K., and Sapkota, K. (2020). The origin of NMDA receptor hypofunction in schizophrenia. Pharmacol. Ther. 205:107426. doi: 10.1016/j.pharmthera.2019.107426

PubMed Abstract | Crossref Full Text | Google Scholar

Neves, D., Salazar, I. L., Almeida, R. D., and Silva, R. M. (2023). Molecular mechanisms of ischemia and glutamate excitotoxicity. Life Sci. 328:121814. doi: 10.1016/j.lfs.2023.121814

PubMed Abstract | Crossref Full Text | Google Scholar

Niethammer, M., Kim, E., and Sheng, M. (1996). Interaction between the C terminus of NMDA receptor subunits and multiple members of the PSD-95 family of membrane-associated guanylate kinases. J. Neurosci. 16, 2157–2163. doi: 10.1523/JNEUROSCI.16-07-02157.1996

PubMed Abstract | Crossref Full Text | Google Scholar

Ondo, W. G., Mejia, N. I., and Hunter, C. B. (2007). A pilot study of the clinical efficacy and safety of memantine for Huntington's disease. Parkinsonism Relat. Disord. 13, 453–454. doi: 10.1016/j.parkreldis.2006.08.005

PubMed Abstract | Crossref Full Text | Google Scholar

Ortega Martínez, E., Morales Hernández, M. E., Castillo-González, J., González-Rey, E., and Ruiz Martínez, M. A. (2024). Dopamine-loaded chitosan-coated solid lipid nanoparticles as a promise nanocarriers to the CNS. Neuropharmacology 249:109871. doi: 10.1016/j.neuropharm.2024.109871

PubMed Abstract | Crossref Full Text | Google Scholar

Paillé, V., Picconi, B., Bagetta, V., Ghiglieri, V., Sgobio, C., Di Filippo, M., et al. (2010). Distinct levels of dopamine denervation differentially alter striatal synaptic plasticity and NMDA receptor subunit composition. J. Neurosci. 30, 14182–14193. doi: 10.1523/JNEUROSCI.2149-10.2010

PubMed Abstract | Crossref Full Text | Google Scholar

Pang, W., and Hu, F. (2021). Cellular and physiological functions of C9ORF72 and implications for ALS/FTD. J. Neurochem. 157, 334–350. doi: 10.1111/jnc.15255

PubMed Abstract | Crossref Full Text | Google Scholar

Paoletti, P. (2011). Molecular basis of NMDA receptor functional diversity. Eur. J. Neurosci. 33, 1351–1365. doi: 10.1111/j.1460-9568.2011.07628.x

Crossref Full Text | Google Scholar

Paoletti, P., Bellone, C., and Zhou, Q. (2013). NMDA receptor subunit diversity: impact on receptor properties, synaptic plasticity and disease. Nat. Rev. Neurosci. 14, 383–400. doi: 10.1038/nrn3504

PubMed Abstract | Crossref Full Text | Google Scholar

Papa, S. M., and Chase, T. N. (1996). Levodopa-induced dyskinesias improved by a glutamate antagonist in parkinsonian monkeys. Ann. Neurol. 39, 574–578. doi: 10.1002/ana.410390505

PubMed Abstract | Crossref Full Text | Google Scholar

Papadia, S., Stevenson, P., Hardingham, N. R., Bading, H., and Hardingham, G. E. (2005). Nuclear Ca2+ and the cAMP response element-binding protein family mediate a late phase of activity-dependent neuroprotection. J. Neurosci. 25, 4279–4287. doi: 10.1523/JNEUROSCI.5019-04.2005

PubMed Abstract | Crossref Full Text | Google Scholar

Park, D. K., Stein, I. S., and Zito, K. (2022). Ion flux-independent NMDA receptor signaling. Neuropharmacology 210:109019. doi: 10.1016/j.neuropharm.2022.109019

PubMed Abstract | Crossref Full Text | Google Scholar

Parsons, M. P., and Raymond, L. A. (2014). Extrasynaptic NMDA receptor involvement in central nervous system disorders. Neuron 82, 279–293. doi: 10.1016/j.neuron.2014.03.030

PubMed Abstract | Crossref Full Text | Google Scholar

Paul, P., and de Belleroche, J. (2014). The role of D-serine and glycine as co-agonists of NMDA receptors in motor neuron degeneration and amyotrophic lateral sclerosis (ALS). Front. Synaptic Neurosci. 6:10. doi: 10.3389/fnsyn.2014.00010

PubMed Abstract | Crossref Full Text | Google Scholar

Paul, P., Murphy, T., Oseni, Z., Sivalokanathan, S., and de Belleroche, J. S. (2014). Pathogenic effects of amyotrophic lateral sclerosis-linked mutation in D-amino acid oxidase are mediated by D-serine. Neurobiol. Aging 35, 876–885. doi: 10.1016/j.neurobiolaging.2013.09.005

PubMed Abstract | Crossref Full Text | Google Scholar

Petralia, R. S., Wang, Y. X., Hua, F., Yi, Z., Zhou, A., Ge, L., et al. (2010). Organization of NMDA receptors at extrasynaptic locations. Neuroscience 167, 68–87. doi: 10.1016/j.neuroscience.2010.01.022

PubMed Abstract | Crossref Full Text | Google Scholar

Picconi, B., Piccoli, G., and Calabresi, P. (2012). Synaptic dysfunction in Parkinson's disease. Adv. Exp. Med. Biol. 970, 553–572. doi: 10.1007/978-3-7091-0932-8_24

PubMed Abstract | Crossref Full Text | Google Scholar

Puspita, L., Chung, S. Y., and Shim, J. W. (2017). Oxidative stress and cellular pathologies in Parkinson's disease. Mol. Brain 10:53. doi: 10.1186/s13041-017-0340-9

PubMed Abstract | Crossref Full Text | Google Scholar

Qin, W., Haroutunian, V., Katsel, P., Cardozo, C. P., Ho, L., Buxbaum, J. D., et al. (2009). PGC-1alpha expression decreases in the Alzheimer disease brain as a function of dementia. Arch. Neurol. 66, 352–361. doi: 10.1001/archneurol.2008.588

PubMed Abstract | Crossref Full Text | Google Scholar

Raïch, I., Lillo, J., Rebassa, J. B., Capó, T., Cordomí, A., Reyes-Resina, I., et al. (2024). Dual role of NMDAR containing NR2A and NR2B subunits in Alzheimer's disease. Int. J. Mol. Sci. 25:757. doi: 10.3390/ijms25094757

PubMed Abstract | Crossref Full Text | Google Scholar

Raymond, L. A., André, V. M., Cepeda, C., Gladding, C. M., Milnerwood, A. J., and Levine, M. S. (2011). Pathophysiology of Huntington's disease: time-dependent alterations in synaptic and receptor function. Neuroscience 198, 252–273. doi: 10.1016/j.neuroscience.2011.08.052

PubMed Abstract | Crossref Full Text | Google Scholar

Raynaud, F., and Marcilhac, A. (2006). Implication of calpain in neuronal apoptosis. A possible regulation of Alzheimer's disease. FEBS J. 273, 3437–3443. doi: 10.1111/j.1742-4658.2006.05352.x

PubMed Abstract | Crossref Full Text | Google Scholar

Reed, T. T. (2011). Lipid peroxidation and neurodegenerative disease. Free Radic. Biol. Med. 51, 1302–1319. doi: 10.1016/j.freeradbiomed.2011.06.027

PubMed Abstract | Crossref Full Text | Google Scholar

Rego, A. C., and Oliveira, C. R. (2003). Mitochondrial dysfunction and reactive oxygen species in excitotoxicity and apoptosis: implications for the pathogenesis of neurodegenerative diseases. Neurochem. Res. 28, 1563–1574. doi: 10.1023/A:1025682611389

PubMed Abstract | Crossref Full Text | Google Scholar

Reisberg, B., Doody, R., Stöffler, A., Schmitt, F., Ferris, S., and Möbius, H. J. (2003). Memantine in moderate-to-severe Alzheimer's disease. N. Engl. J. Med. 348, 1333–1341. doi: 10.1056/NEJMoa013128

PubMed Abstract | Crossref Full Text | Google Scholar

Robinson, S. R. (2000). Neuronal expression of glutamine synthetase in Alzheimer's disease indicates a profound impairment of metabolic interactions with astrocytes. Neurochem. Int. 36, 471–482. doi: 10.1016/S0197-0186(99)00150-3

PubMed Abstract | Crossref Full Text | Google Scholar

Rodriguez, M. C., Obeso, J. A., and Olanow, C. W. (1998). Subthalamic nucleus-mediated excitotoxicity in Parkinson's disease: a target for neuroprotection. Ann. Neurol. 44, S175–S188. doi: 10.1002/ana.410440726

PubMed Abstract | Crossref Full Text | Google Scholar

Rodriguez-Perdigon, M., Tordera, R. M., Gil-Bea, F. J., Gerenu, G., Ramirez, M. J., and Solas, M. (2016). Down-regulation of glutamatergic terminals (VGLUT1) driven by aβ in Alzheimer's disease. Hippocampus 26, 1303–1312. doi: 10.1002/hipo.22607

PubMed Abstract | Crossref Full Text | Google Scholar

Rosa, E., and Fahnestock, M. (2015). CREB expression mediates amyloid β-induced basal BDNF downregulation. Neurobiol. Aging 36, 2406–2413. doi: 10.1016/j.neurobiolaging.2015.04.014

PubMed Abstract | Crossref Full Text | Google Scholar

Rothman, S. M., and Olney, J. W. (1987). Excitotoxity and the NMDA receptor. Trends Neurosci. 10, 299–302. doi: 10.1016/0166-2236(87)90177-9

Crossref Full Text | Google Scholar

Rothman, S. M., and Olney, J. W. (1995). Excitotoxicity and the NMDA receptor - still lethal after eight years. Trends Neurosci. 18, 57–58. doi: 10.1016/0166-2236(95)93869-y

PubMed Abstract | Crossref Full Text | Google Scholar

Rothstein, J. D., Tsai, G., Kuncl, R. W., Clawson, L., Cornblath, D. R., Drachman, D. B., et al. (1990). Abnormal excitatory amino acid metabolism in amyotrophic lateral sclerosis. Ann. Neurol. 28, 18–25. doi: 10.1002/ana.410280106

PubMed Abstract | Crossref Full Text | Google Scholar

Rothstein, J. D., Van Kammen, M., Levey, A. I., Martin, L. J., and Kuncl, R. W. (1995). Selective loss of glial glutamate transporter GLT-1 in amyotrophic lateral sclerosis. Ann. Neurol. 38, 73–84. doi: 10.1002/ana.410380114

PubMed Abstract | Crossref Full Text | Google Scholar

Sanz-Clemente, A., Nicoll, R. A., and Roche, K. W. (2013). Diversity in NMDA receptor composition: many regulators, many consequences. Neuroscientist 19, 62–75. doi: 10.1177/1073858411435129

PubMed Abstract | Crossref Full Text | Google Scholar

Sasabe, J., Miyoshi, Y., Suzuki, M., Mita, M., Konno, R., Matsuoka, M., et al. (2012). D-amino acid oxidase controls motoneuron degeneration through D-serine. Proc. Natl. Acad. Sci. USA 109, 627–632. doi: 10.1073/pnas.1114639109

PubMed Abstract | Crossref Full Text | Google Scholar

Scheltens, P., De Strooper, B., Kivipelto, M., Holstege, H., Chételat, G., Teunissen, C. E., et al. (2021). Alzheimer's disease. Lancet 397, 1577–1590. doi: 10.1016/S0140-6736(20)32205-4

PubMed Abstract | Crossref Full Text | Google Scholar

Schuch, F. B., Vancampfort, D., Richards, J., Rosenbaum, S., Ward, P. B., and Stubbs, B. (2016). Exercise as a treatment for depression: a meta-analysis adjusting for publication bias. J. Psychiatr. Res. 77, 42–51. doi: 10.1016/j.jpsychires.2016.02.023

PubMed Abstract | Crossref Full Text | Google Scholar

Scimemi, A., Meabon, J. S., Woltjer, R. L., Sullivan, J. M., Diamond, J. S., and Cook, D. G. (2013). Amyloid-β1-42 slows clearance of synaptically released glutamate by mislocalizing astrocytic GLT-1. J. Neurosci. 33, 5312–5318. doi: 10.1523/JNEUROSCI.5274-12.2013

PubMed Abstract | Crossref Full Text | Google Scholar

Sepers, M. D., and Raymond, L. A. (2014). Mechanisms of synaptic dysfunction and excitotoxicity in Huntington's disease. Drug Discov. Today 19, 990–996. doi: 10.1016/j.drudis.2014.02.006

PubMed Abstract | Crossref Full Text | Google Scholar

Shi, Y., Lin, S., Staats, K. A., Li, Y., Chang, W. H., Hung, S. T., et al. (2018). Haploinsufficiency leads to neurodegeneration in C9ORF72 ALS/FTD human induced motor neurons. Nat. Med. 24, 313–325. doi: 10.1038/nm.4490

PubMed Abstract | Crossref Full Text | Google Scholar

Singh, A., Kukreti, R., Saso, L., and Kukreti, S. (2019). Oxidative stress: a key modulator in neurodegenerative diseases. Molecules 24:583. doi: 10.3390/molecules24081583

PubMed Abstract | Crossref Full Text | Google Scholar

Sitcheran, R., Gupta, P., Fisher, P. B., and Baldwin, A. S. (2005). Positive and negative regulation of EAAT2 by NF-kappaB: a role for N-myc in TNFalpha-controlled repression. EMBO J. 24, 510–520. doi: 10.1038/sj.emboj.7600555

PubMed Abstract | Crossref Full Text | Google Scholar

Snow, W. M., and Albensi, B. C. (2016). Neuronal gene targets of NF-κB and their dysregulation in Alzheimer's disease. Front. Mol. Neurosci. 9:118. doi: 10.3389/fnmol.2016.00118

PubMed Abstract | Crossref Full Text | Google Scholar

Spalloni, A., Nutini, M., and Longone, P. (2013). Role of the N-methyl-d-aspartate receptors complex in amyotrophic lateral sclerosis. Biochim. Biophys. Acta 1832, 312–322. doi: 10.1016/j.bbadis.2012.11.013

PubMed Abstract | Crossref Full Text | Google Scholar

Stark, D. T., and Bazan, N. G. (2011). Synaptic and extrasynaptic NMDA receptors differentially modulate neuronal cyclooxygenase-2 function, lipid peroxidation, and neuroprotection. J. Neurosci. 31, 13710–13721. doi: 10.1523/JNEUROSCI.3544-11.2011

PubMed Abstract | Crossref Full Text | Google Scholar

Su, Z. Z., Leszczyniecka, M., Kang, D. C., Sarkar, D., Chao, W., Volsky, D. J., et al. (2003). Insights into glutamate transport regulation in human astrocytes: cloning of the promoter for excitatory amino acid transporter 2 (EAAT2). Proc. Natl. Acad. Sci. USA 100, 1955–1960. doi: 10.1073/pnas.0136555100

PubMed Abstract | Crossref Full Text | Google Scholar

Suárez, F., Zhao, Q., Monaghan, D. T., Jane, D. E., Jones, S., and Gibb, A. J. (2010). Functional heterogeneity of NMDA receptors in rat substantia nigra pars compacta and reticulata neurones. Eur. J. Neurosci. 32, 359–367. doi: 10.1111/j.1460-9568.2010.07298.x

PubMed Abstract | Crossref Full Text | Google Scholar

Sulzer, D., and Surmeier, D. J. (2013). Neuronal vulnerability, pathogenesis, and Parkinson's disease. Mov. Disord. 28, 41–50. doi: 10.1002/mds.25095

PubMed Abstract | Crossref Full Text | Google Scholar

Sun, E., Motolani, A., Campos, L., and Lu, T. (2022). The pivotal role of NF-kB in the pathogenesis and therapeutics of Alzheimer's disease. Int. J. Mol. Sci. 23:72. doi: 10.3390/ijms23168972

PubMed Abstract | Crossref Full Text | Google Scholar

Sun, Y., Savanenin, A., Reddy, P. H., and Liu, Y. F. (2001). Polyglutamine-expanded huntingtin promotes sensitization of N-methyl-d-aspartate receptors via post-synaptic density 95*. J. Biol. Chem. 276, 24713–24718. doi: 10.1074/jbc.M103501200

PubMed Abstract | Crossref Full Text | Google Scholar

Sun, G. Y., Xu, J., Jensen, M. D., and Simonyi, A. (2004). Phospholipase A2 in the central nervous system: implications for neurodegenerative diseases. J. Lipid Res. 45, 205–213. doi: 10.1194/jlr.R300016-JLR200

PubMed Abstract | Crossref Full Text | Google Scholar

Swanger, S. A., Vance, K. M., Pare, J. F., Sotty, F., Fog, K., Smith, Y., et al. (2015). NMDA receptors containing the GluN2D subunit control neuronal function in the subthalamic nucleus. J. Neurosci. 35, 15971–15983. doi: 10.1523/JNEUROSCI.1702-15.2015

PubMed Abstract | Crossref Full Text | Google Scholar

Sweeney, G., and Song, J. (2016). The association between PGC-1α and Alzheimer's disease. Anat. Cell Biol. 49, 1–6. doi: 10.5115/acb.2016.49.1.1

PubMed Abstract | Crossref Full Text | Google Scholar

Szydlowska, K., and Tymianski, M. (2010). Calcium, ischemia and excitotoxicity. Cell Calcium 47, 122–129. doi: 10.1016/j.ceca.2010.01.003

PubMed Abstract | Crossref Full Text | Google Scholar

Tamburri, A., Dudilot, A., Licea, S., Bourgeois, C., and Boehm, J. (2013). NMDA-receptor activation but not ion flux is required for amyloid-beta induced synaptic depression. PLoS One 8:e65350. doi: 10.1371/journal.pone.0065350

PubMed Abstract | Crossref Full Text | Google Scholar

Tao, W. Y., Yu, L. J., Jiang, S., Cao, X., Chen, J., Bao, X. Y., et al. (2020). Neuroprotective effects of ZL006 in aβ(1-42)-treated neuronal cells. Neural Regen. Res. 15, 2296–2305. doi: 10.4103/1673-5374.285006

PubMed Abstract | Crossref Full Text | Google Scholar

Tian, Z., Lu, X. T., Jiang, X., and Tian, J. (2023). Bryostatin-1: a promising compound for neurological disorders. Front. Pharmacol. 14:1187411. doi: 10.3389/fphar.2023.1187411

PubMed Abstract | Crossref Full Text | Google Scholar

Tripathi, M. K., Kartawy, M., and Amal, H. (2020). The role of nitric oxide in brain disorders: autism spectrum disorder and other psychiatric, neurological, and neurodegenerative disorders. Redox Biol. 34:101567. doi: 10.1016/j.redox.2020.101567

PubMed Abstract | Crossref Full Text | Google Scholar

Trudler, D., Sanz-Blasco, S., Eisele, Y. S., Ghatak, S., Bodhinathan, K., Akhtar, M. W., et al. (2021). Α-Synuclein oligomers induce glutamate release from astrocytes and excessive Extrasynaptic NMDAR activity in neurons, thus contributing to synapse loss. J. Neurosci. 41, 2264–2273. doi: 10.1523/JNEUROSCI.1871-20.2020

PubMed Abstract | Crossref Full Text | Google Scholar

Tu, Y. C., and Kuo, C. C. (2015). The differential contribution of GluN1 and GluN2 to the gating operation of the NMDA receptor channel. Pflugers Arch. 467, 1899–1917. doi: 10.1007/s00424-014-1630-z

PubMed Abstract | Crossref Full Text | Google Scholar

Ugale, V., Deshmukh, R., Lokwani, D., Narayana Reddy, P., Khadse, S., Chaudhari, P., et al. (2024). GluN2B subunit selective N-methyl-D-aspartate receptor ligands: democratizing recent progress to assist the development of novel neurotherapeutics. Mol. Divers. 28, 1765–1792. doi: 10.1007/s11030-023-10656-0

PubMed Abstract | Crossref Full Text | Google Scholar

Um, J. W., Kaufman, A. C., Kostylev, M., Heiss, J. K., Stagi, M., Takahashi, H., et al. (2013). Metabotropic glutamate receptor 5 is a coreceptor for Alzheimer aβ oligomer bound to cellular prion protein. Neuron 79, 887–902. doi: 10.1016/j.neuron.2013.06.036

PubMed Abstract | Crossref Full Text | Google Scholar

Um, J. W., Nygaard, H. B., Heiss, J. K., Kostylev, M. A., Stagi, M., Vortmeyer, A., et al. (2012). Alzheimer amyloid-β oligomer bound to postsynaptic prion protein activates Fyn to impair neurons. Nat. Neurosci. 15, 1227–1235. doi: 10.1038/nn.3178

PubMed Abstract | Crossref Full Text | Google Scholar

Van Den Bosch, L., Van Damme, P., Bogaert, E., and Robberecht, W. (2006). The role of excitotoxicity in the pathogenesis of amyotrophic lateral sclerosis. Biochim. Biophys. Acta 1762, 1068–1082. doi: 10.1016/j.bbadis.2006.05.002

PubMed Abstract | Crossref Full Text | Google Scholar

Vico Varela, E., Etter, G., and Williams, S. (2019). Excitatory-inhibitory imbalance in Alzheimer's disease and therapeutic significance. Neurobiol. Dis. 127, 605–615. doi: 10.1016/j.nbd.2019.04.010

PubMed Abstract | Crossref Full Text | Google Scholar

Vyklicky, V., Korinek, M., Smejkalova, T., Balik, A., Krausova, B., Kaniakova, M., et al. (2014). Structure, function, and pharmacology of NMDA receptor channels. Physiol. Res. 63, S191–S203. doi: 10.33549/physiolres.932678

PubMed Abstract | Crossref Full Text | Google Scholar

Walker, F. O. (2007). Huntington's disease. Lancet 369, 218–228. doi: 10.1016/S0140-6736(07)60111-1

PubMed Abstract | Crossref Full Text | Google Scholar

Wang, R., and Reddy, P. H. (2017). Role of glutamate and NMDA receptors in Alzheimer's disease. J. Alzheimers Dis. 57, 1041–1048. doi: 10.3233/JAD-160763

PubMed Abstract | Crossref Full Text | Google Scholar

Wang, S., Shi, X., Li, H., Pang, P., Pei, L., Shen, H., et al. (2017). DAPK1 signaling pathways in stroke: from mechanisms to therapies. Mol. Neurobiol. 54, 4716–4722. doi: 10.1007/s12035-016-0008-y

PubMed Abstract | Crossref Full Text | Google Scholar

Wang, Q., Walsh, D. M., Rowan, M. J., Selkoe, D. J., and Anwyl, R. (2004). 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. 24, 3370–3378. doi: 10.1523/JNEUROSCI.1633-03.2004

PubMed Abstract | Crossref Full Text | Google Scholar

Wang, R., and Zhang, D. (2005). Memantine prolongs survival in an amyotrophic lateral sclerosis mouse model. Eur. J. Neurosci. 22, 2376–2380. doi: 10.1111/j.1460-9568.2005.04431.x

PubMed Abstract | Crossref Full Text | Google Scholar

Wang, Z. C., Zhao, J., and Li, S. (2013). Dysregulation of synaptic and extrasynaptic N-methyl-D-aspartate receptors induced by amyloid-β. Neurosci. Bull. 29, 752–760. doi: 10.1007/s12264-013-1383-2

PubMed Abstract | Crossref Full Text | Google Scholar

Wareski, P., Vaarmann, A., Choubey, V., Safiulina, D., Liiv, J., Kuum, M., et al. (2009). PGC-1{alpha} and PGC-1{beta} regulate mitochondrial density in neurons. J. Biol. Chem. 284, 21379–21385. doi: 10.1074/jbc.M109.018911

PubMed Abstract | Crossref Full Text | Google Scholar

Watanabe, M., Mishina, M., and Inoue, Y. (1994). Distinct spatiotemporal expressions of five NMDA receptor channel subunit mRNAs in the cerebellum. J. Comp. Neurol. 343, 513–519. doi: 10.1002/cne.903430402

PubMed Abstract | Crossref Full Text | Google Scholar

Wesseling, J. F., and Pérez-Otaño, I. (2015). Modulation of GluN3A expression in Huntington disease: a new n-methyl-D-aspartate receptor-based therapeutic approach? JAMA Neurol. 72, 468–473. doi: 10.1001/jamaneurol.2014.3953

PubMed Abstract | Crossref Full Text | Google Scholar

Wyllie, D. J., Livesey, M. R., and Hardingham, G. E. (2013). Influence of GluN2 subunit identity on NMDA receptor function. Neuropharmacology 74, 4–17. doi: 10.1016/j.neuropharm.2013.01.016

PubMed Abstract | Crossref Full Text | Google Scholar

Xia, P., Chen, H. S., Zhang, D., and Lipton, S. A. (2010). Memantine preferentially blocks extrasynaptic over synaptic NMDA receptor currents in hippocampal autapses. J. Neurosci. 30, 11246–11250. doi: 10.1523/JNEUROSCI.2488-10.2010

PubMed Abstract | Crossref Full Text | Google Scholar

Xu, Y., Yan, J., Zhou, P., Li, J., Gao, H., Xia, Y., et al. (2012). Neurotransmitter receptors and cognitive dysfunction in Alzheimer's disease and Parkinson's disease. Prog. Neurobiol. 97, 1–13. doi: 10.1016/j.pneurobio.2012.02.002

PubMed Abstract | Crossref Full Text | Google Scholar

Yukawa, T., Yamashita, T., Imaeda, T., Kakei, H., Hashizume, S., Nakamura, M., et al. (2023). Design and synthesis of 6-methylpyridin-2-one derivatives as novel and potent GluN2A positive allosteric modulators for the treatment of cognitive impairment. Bioorg. Med. Chem. 79:117150. doi: 10.1016/j.bmc.2022.117150

PubMed Abstract | Crossref Full Text | Google Scholar

Zhang, X., and Chergui, K. (2015). Dopamine depletion of the striatum causes a cell-type specific reorganization of GluN2B- and GluN2D-containing NMDA receptors. Neuropharmacology 92, 108–115. doi: 10.1016/j.neuropharm.2015.01.007

PubMed Abstract | Crossref Full Text | Google Scholar

Zhang, F., Liu, M., Tuo, J., Zhang, L., Zhang, J., Yu, C., et al. (2023). Levodopa-induced dyskinesia: interplay between the N-methyl-D-aspartic acid receptor and neuroinflammation. Front. Immunol. 14:1253273. doi: 10.3389/fimmu.2023.1253273

PubMed Abstract | Crossref Full Text | Google Scholar

Zhang, C., Liu, X., Xu, H., Hu, G., Zhang, X., Xie, Z., et al. (2020). Protopanaxadiol ginsenoside Rd protects against NMDA receptor-mediated excitotoxicity by attenuating calcineurin-regulated DAPK1 activity. Sci. Rep. 10:8078. doi: 10.1038/s41598-020-64738-2

PubMed Abstract | Crossref Full Text | Google Scholar

Glossary

NMDAR - N-methyl-D-aspartate receptor

ALS - Amyotrophic Lateral Sclerosis

PD - Parkinson’s Disease

HD - Huntington’s Disease

AD - Alzheimer’s Disease

LTP - Long-term Potentiation

LTD - Long-term Depression

PSD - Postsynaptic Density

AMPAR - Amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor

CNS - Central Nervous System

EAAT2 - Excitatory Amino Acid Transporter 2

CREB - cAMP Response Element-binding Protein

BDNF - Brain-derived neurotrophic factor

ROS - Reactive Oxygen Species

nNOS - neuronal Nitric Oxide Synthase

PLA2 - Phospholipase A2

PUFAs - Polyunsaturated Fatty Acids

AA - Arachidonic Acid

4-HNE - 4-hydroxynonenal

MDA - Malondialdehyde

D-APV - D-2-Amino-5-phosphonopentanoic acid

7CK - 7 chloro-kynurenate (7CK)

SNc - Substantia Nigra Pars Compacta

STN - Subthalamic Nucleus

LID - Levodopa-induced Dyskinesia

DAPK1 - Death-associated Protein Kinase 1

NF-κB - Nuclear Factor Kappa-Light-Chain-Enhancer of Activated B Cells

HTT - huntingtin

mHTT - mutant Huntingtin

MSNs - Medium Spiny Neurons

PrPc - Cellular Prion Protein

mGluR5 - Metabotropic Glutamate Receptor 5

- Amyloid-β

GS - Glutamine Synthetase

Keywords: NMDA receptors (N-methyl-D-aspartate receptors), neurodegenerative disease, excitotoxicity, glutamate, Alzheimer’s disease (AD), Huntington’s disease (HD), Parkinson’s disease (PD), amyotrophic lateral sclerosis (ALS)

Citation: Zhang K, Wen M, Nan X, Zhao S, Li H, Ai Y and Zhu H (2025) NMDA receptors in neurodegenerative diseases: mechanisms and emerging therapeutic strategies. Front. Aging Neurosci. 17:1604378. doi: 10.3389/fnagi.2025.1604378

Received: 01 April 2025; Accepted: 30 June 2025;
Published: 24 July 2025.

Edited by:

Jean Maurice Delabar, Centre National de la Recherche Scientifique (CNRS), France

Reviewed by:

Consuelo Morgado-Valle, Universidad Veracruzana, Mexico
Md. Jakaria, Purdue University, United States

Copyright © 2025 Zhang, Wen, Nan, Zhao, Li, Ai and Zhu. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Houze Zhu, emh1aHotbXJyaWdodEAxNjMuY29t; Yanping AiMzI3NDE1NjhAcXEuY29t; Hao Li, bGloYW9jaG5AaHVzdC5lZHUuY24=

These authors have contributed equally to this work

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.