Next Article in Journal
Systemic Chronic Inflammation: Integrative Strategies for Health Improvement and Prevention of Inflammatory Receptor Overexpression
Previous Article in Journal
RNase P-Associated External Guide Sequences Inhibit HIV-1 Infection by Shutting Down Human CCR5 Expression
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Glutamate Receptor Signaling in Retina Müller Cells: Plausible Role in Neurodegeneration

by
Bolaji Oyetayo
1,
Yurixy Mendoza-Silva
1,
Temitayo Subair
1,
Luisa C Hernández-Kelly
1,
Marie-Paule Felder-Schmittbuhl
2,
Tatiana N. Olivares-Bañuelos
3 and
Arturo Ortega
1,*
1
Department of Toxicology, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Av. IPN 2508, Mexico City 07360, Mexico
2
Centre National de la Recherche Scientifique, Institute of Cellular and Integrative Neurosciences (UPR 3212), Université de Strasbourg, 67081 Strasbourg, France
3
Instituto de Investigaciones Oceanológicas, Universidad Autónoma de Baja California, Carretera Ensenada-Tijuana No. 3917, Fracc. Playitas, Ensenada 22860, Mexico
*
Author to whom correspondence should be addressed.
Receptors 2025, 4(1), 4; https://doi.org/10.3390/receptors4010004
Submission received: 26 November 2024 / Revised: 31 January 2025 / Accepted: 19 February 2025 / Published: 26 February 2025

Abstract

:
The retinal network relies on glutamate, the primary excitatory neurotransmitter involved in the visual cycle. Glutamate transactions are carried out by an array of distinct receptors and transporters distributed across both pre- and post-synaptic neurons and Müller radial glial cells. Glutamate receptors are broadly divided into two types: ionotropic and metabotropic receptors that differ in their molecular architecture and signaling properties. Within the retina, Müller glia cells span across its entire layers and possess specialized features that enable them to regulate glutamate extracellular levels and thus, its neuronal availability. In order to prevent an excitotoxic insult, retina extracellular glutamate levels have to be tightly regulated through uptake, predominantly into Müller glial cells, by a family of Na+-dependent glutamate transporters known as excitatory amino acid transporters. An exquisite interplay between glutamate receptor signaling and glutamate transporter expression and function is fundamental for the integrity and proper function of the retina. This review examines our current understanding of the impact of Müller glial glutamate signaling on glia/neuronal coupling.

1. Introduction

The retina is a delicate and vital part of the central nervous system (CNS) which contributes to our understanding of the brain with its surprisingly intricate cellular architecture [1]. This structure is a complex mosaic encompassing five distinct classes of specialized neurons dedicated to processing information received by the eye [2,3,4]. Retinal neurons—namely photoreceptors, bipolar cells, ganglion cells, horizontal cells, and amacrine cells—exhibit a characteristic distribution across the retinal layers [1,2]. Retinal neurons are interconnected through the vertical and horizontal pathways collaboratively contributing to the processing of visual images and the subsequent transmission of this information to the brain via the optic nerve [3]. The horizontal pathway involves lateral connections between horizontal cells and amacrine cells, facilitating feedback signals between photoreceptors and bipolar cells (for horizontal cells) and between bipolar cells and retinal ganglion cells (RGCs) (for amacrine cells) [5]. Conversely, the vertical pathway initiates the transduction of light signals by photoreceptors and bipolar cells, culminating in the transmission to the brain through the axons of RGCs (Figure 1). The excitatory neurotransmitter glutamate (Glu) is the most important signal in this pathway. Across retinal neurons, various neurotransmitter receptors are diversely expressed and this contributes significantly to functional diversity, forming the foundation for multiple processing pathways of visual information. Two primary classes of receptors mediate the effects of Glu: ionotropic receptors (GRI) and metabotropic receptors (GRM) [6]. A plethora of factors regulate the activities of these receptors such as subunit composition, density, receptor affinity, and localization at the synapse [7].
The retina also comprises non-neuronal cells such as astrocytes, microglia, and Müller cells (MCs) that support retinal neurons. Astrocytes are confined to the innermost layers of vascularized retinas. In contrast, MCs are radially shaped glial cells that extend across the entire thickness of the retinal neurons establishing close connections and contributing significantly to the preservation of tissue structure, homeostasis, and light transmission (Figure 1) [8,9]. MCs play a crucial role in sustaining the functionality and metabolism of retinal neurons, actively contributing to both normal retinal function and the processes associated with retinal degeneration [10]. MCs constitute a now accepted, indispensable component of a robust and optimally functioning retina [11]. Disruptions in these cells inevitably perturb cellular communication within the retina, compromising its proper function. The precision of metabolic processes in MCs can be quantified and is an actively explored area. Investigating the factors triggering metabolic changes in disease and understanding the impact of these changes on MCs’ ability to support neuronal function is crucial for comprehending the progression of retinal disease and fundamental for designing therapeutics compatible with the diseased retina. Despite their significance, MCs remain a relatively understudied cell type despite their role in excitotoxicity and neurodegeneration. In this contribution, we seek to provide a comprehensive overview of Glu receptor expression and signaling in MCs and delineate the MC’s role in the prevention of excitotoxicity events occurring in the retina.

2. Glu-Mediated Neurotransmission in the Retina

In the retina, the process whereby light energy is converted into visual information is known as phototransduction; this activity is modulated by a diverse array of retinal cells, neurotransmitters, transporters, and receptors [12]. The phototransduction process is made up of four sequential steps. Photoreceptors are the first point of contact with light rays and they are made of two types: rods and cones. Photoreceptor cells possess different features such as chromatic sensitivity and relative abundance. Moreover, the photoreceptors’ relative abundance and chromatic sensitivity, rods and cones play a critical role in light sensitivity. An increase in light reduces the sensitivity of rods thereby activating the cones for the photopic vision. Yin et al. [13] showed that a mixture of signals between the rods and cones occurs as illumination increases from nearly 100% of rods in the low-mesopic range to about 20% in the mid-level photopic range. The activation of rod signals in the low-mesopic range to the relatively high photopic range in the cone visual pathway experiment conducted in retinal ganglion cells was shown to be an adaptive mechanism for varying illumination signals hence emphasizing the critical role of photoreceptors in the visual pathway [14].
In the light cycle, light absorption occurs when photons hit the photoreceptors and it is captured by the visual photopigments (rhodopsin) present in rods and cones [15,16]. In response to light conditions, photons are absorbed by rhodopsin and cis-retinal converted into an isomerized form (all-trans-retinal). The conformational change allows the disintegration of the opsin from the retinal, adopting an unstable conformation called “metarhodopsin”. cGMP is hydrolyzed by the transducin α subunit, a G-protein, activated by metarhodopsin. Once activated, it stimulates a cGMP phosphodiesterase (PDE) [17,18] leading to the closure of Na+ channels thereby hyperpolarizing the cells [16]. This leads to a shift in the membrane potential from −40 mV to −75 mV [16]. Due to this, K+ channels open and Ca2+ channels close [19], causing the vesicles loaded with glutamate (Glu) to be trapped within the presynaptic axon terminals [20], reducing the amount of Glu released [21]. This amino acid is widely considered the primary excitatory neurotransmitter in the retina [22] and plays a crucial role in the forward transmission of visual signals among photoreceptors, bipolar and ganglion cells [23,24].
Under dark conditions, photoreceptors are depolarized due to the opening of cGMP-dependent Na+ channels. The increase in intracellular Ca2+ concentrations causes the synaptic vesicles loaded with Glu to move to the active zone and fuse with the membrane, then Glu is released into the synaptic cleft [20,25,26].
Consequently, bipolar cells serve as the site for the separation of ON and OFF signaling [16]. OFF bipolar cells, equipped with ionotropic Glu receptors, depolarize in the dark in response to the Glu released by photoreceptors [27,28]. On the other hand, ON bipolar cells express metabotropic Glu receptors, specifically mGluR6, on their dendrites [29]. The activation of mGluR6, linked to the G protein Go, triggers a downstream second-messenger system, resulting in hyperpolarization of the cell following Glu binding [30,31]. Therefore, ON bipolar cells exhibit hyperpolarization in the dark, contrasting with the photoreceptors and OFF bipolar cells [32]. Photoreceptors and bipolar cells are unique in their lack of action potential generation and response to light through graded potentials, thereby influencing the release of Glu [33]. In the visual system, synaptic ribbons are also present in the output synapses of second-order retinal bipolar neurons, which, like retinal photoreceptors, auditory hair cells, and pineal photoreceptors, signal through graded changes in membrane potential [34].
Physiological investigations with isolated cells reveal that only micromolar levels of Glu are necessary to activate Glu receptors [35,36]. Consequently, the quantity of Glu released into the synaptic cleft exceeds the concentration needed to activate most postsynaptic receptors by several orders of magnitude. To prevent cell death, high-affinity Glu transporters known as excitatory amino acid transporters (EAATs) located on adjacent neurons and surrounding MCs swiftly eliminate Glu from the synaptic cleft [37]. Five cloned Glu transporters, namely EAAT-1 (GLAST), EAAT-2 (Glt-1), EAAT-3 (EAAC-1), EAAT-4, and EAAT-5, play pivotal roles in this process [38,39,40]. Neuronal Glu transporters include the excitatory amino acid carrier (EAAC-1), EAAT-4 and EAAT-5 [41]. EAAC-1 is predominantly found in amacrine, ganglion, and horizontal cells. EAAT-4 has been identified in retinal astrocytes and the retinal pigment epithelium [42,43]. EAAT-5 is well expressed in photoreceptors, bipolar, and amacrine cells and is characterized by low Glu transport activity and significant anion conductance, functioning primarily as a Glu-activated chloride channel to modulate neuronal excitability [44]. Notably, a splice variant of Glt-1/EAAT-2, known as Glt-1c, is selectively expressed in photoreceptor axon terminals [45].
Distinct from both iGluRs and mGluRs, Glu transporters exhibit unique pharmacological characteristics. Substrates for the transporters include L-Glu, L-aspartate (Asp), D-Asp [46,47], and L-Homocysteate (L-HCA) [48]. While Glu receptor agonists and antagonists are not recognized [49,50]. Upon uptake, Glu transporters incorporate Glu into MCs, concomitant with the co-transport of three Na+ [51] and the antiport of one K+ along with either one OH or one HCO3− [41,52]. This process generates a net positive inward current due to excess Na+, fueling the transporter [53,54,55].

3. Role of Müller Glial Cells in the Retina

Glial cells support the survival and differentiation of neurons by leading developing neurons and their axons to their targets, constituting an essential component of the blood-brain barrier and contributing to the regulation of extracellular ions and neurotransmitter concentrations [56]. Following the completion of neuronal migration, radial glia cells undergo astrocytic conversion, transforming into multipolar astrocytes [57,58]. Notably, exceptions to this astrocytic conversion occur in the cerebellum and retina, where Bergmann and Müller’s glia persist even into adulthood. MCs are the principal radial glial cells within the inner vertebrate retina, exhibiting a distinct cylindrical, fiber-like morphology, originally termed the ‘Müller radial fibers’ [8].
MCs serve as both anatomical and functional bridges connecting retinal neurons with the compartments essential for molecular exchange, including blood vessels, the vitreous chamber, and the subretinal space. MCs in the retina are highly organized and positioned to enable the performance of a diverse array of physiological functions essential for modulating signaling and impacting the survival of retinal neurons [10]. MCs process enwrap synapses and establish connections with blood vessels. MCs are readily distinguishable by their peculiar morphology, featuring a well-developed endoplasmic reticulum, significant mitochondria observed at the end feet, and varying amounts of glycogen granules [59]. In living retinal preparations, MCs can be labeled by brief exposure to dyes like Mito tracker orange or Cell Tracker green. In culture, MCs nuclei can be distinguished as the largest among various retinal cell types [60]. Nuclei staining is achieved with ethidium bromide, diamidino yellow, or chromomycin A3, which selectively penetrates MCs, displaying a pronounced affinity for nucleic acids [61]. Immunohistochemically, the expression of specific protein markers is confined to astrocytes and MCs, such as vimentin and glial fibrillary acidic protein (GFAP) [62], or selectively to MCs, including the cellular retinaldehyde-binding protein (CRALBP), Glu/Asp transporter (GLAST), and GS [63]. However, MCs have been shown to express GFAP in response to retinal diseases and injury [64].
MCs express a diverse array of receptors, encompassing those for amino acids, catecholamines, neuroactive peptides, hormones, and growth factors [65,66,67]. These receptors in turn exist as co-assemblies of different subunits leading to variations in signal transduction [67]. The activation of receptors in glial cells during neuronal activity suggests the potential involvement of glia in information processing, plasticity, or pathological states [57]. MCs protect the retinal neurons due to their unique properties which involve the secretion of neurotrophic factors, timely Glu uptake, recycling, and the release of antioxidants. MCs contribute to the reduced glutathione pool utilized in neurons, acting as a scavenger for free radicals and reactive oxygen compounds. Specialized high-affinity membrane Glu transporters (EAATs) are vital in preventing neurotoxicity by ensuring the prompt clearance of synaptic glutamate [55]. EAATs are expressed in both neurons and MCs. However, the synaptic responses of RGCs are influenced by the Glu transporters of MCs, as opposed to neuronal transporters. The predominant removal of Glu occurs through uptake by MCs, in which GLAST and Glu transporter 1 (Glt-1) play crucial roles.
Glu metabolism within MCs is connected to its nutritional functions [9]. MCs play a crucial role in generating substrates essential for the oxidative metabolism of photoreceptors and neurons, including but not limited to Gln, lactate, pyruvate, alanine, and α-ketoglutarate [68]. These produced substrates serve as vital energy sources for photoreceptors and neurons during periods of metabolic stress, such as in darkness. Additionally, Glu contributes to forming energy substrates through alternative metabolic pathways, including pyruvate transamination [69]. Within the retina, the predominant site of glucose uptake and metabolism is the inner processes of MCs, specifically localized to the inner plexiform and ganglion cell layers, where glutamatergic signaling is important [70]. Experimental inhibition of glial Glu uptake has demonstrated that even low extracellular Glu concentrations are neurotoxic [71]. Changes in the activity of glial Glu transporters may also contribute to regulating the glial support of neuronal signal transmission from photoreceptors to RGCs [9]. Through their involvement in Glu uptake, MCs contribute to establishing the signal-to-noise ratio of synaptic transmission and the spatial resolution of light-induced signaling [72]. Our lab and others have shown that besides Glu transporters’ role in rapidly removing the amino acid from the synaptic cleft, EAATs also serve as an important signaling molecule involved in neuronal/glia coupling. EAATs are differentially expressed, which strongly suggests that each of these transporters is under a specific transcriptional regulation. For a more detailed description of Glu transporter’s expression, regulation and signaling see these recent reviews [41,73,74,75].
In addition to decreasing extra synaptic Glu levels, thereby preventing spillage to nearby synapses, MCs uniquely harbor glutamine synthetase (GS), an enzyme crucial for converting Glu into Gln [76,77]. The rapid activity of the GS enzyme could be responsible for the resistant properties shown by MCs to excitotoxic Glu levels as described in chick cerebellum-derived Bergmann glia cell (BGC) [78]. However, studies have shown that prolonged exposure to Glu can also induce changes in glial cell morphology without resulting in cell death. Observed alterations include a collapse of the cytoskeleton and a subsequent transition from an epithelioid to a retracted morphology [79].

4. Glu Receptor Classification

Glu receptors fall into two main classes: metabotropic and ionotropic, which are categorized into distinct subtypes and subgroups (Figure 2) based on their affinity to specific Glu analogs, subsequent signal induction mechanisms, sequence homology, and structure [80]. While many Glu receptor agonists and antagonists share structural similarities with Glu, this is not universally true for all compounds. Reviews by Reiner et al. [81] and others [82,83] describe Glu receptors’ pharmacological and molecular properties extensively.

5. Ionotropic Glu Receptor (GRI)

Ionotropic Glu receptors are integral membrane proteins characterized by the assembly of four different subunits (tetramers), each comprising over 900 residues, forming a central ion channel pore [83]. The GRI family are ligand-gated cation channels that account for most of the fast excitatory synaptic transmission in the CNS. It includes α-amino-3-hydroxy-5-methyl-4-isoxazolepropionate (GRIA), kainic acid (GRIK), and N-methyl-D-Aspartate receptors (GRIN) [84,85]. GRI are characterized by sequence similarity which suggests a common architectural framework. GRIA and GRIK subunits typically form functional homotetramers or heterotetramers, although NMDA receptors are predominantly heterotetramers [86]. Notably, across all GRI subtypes, the binding sites for pore blockers are consistently present within the transmembrane ion channel, emphasizing a shared pharmacological aspect critical to their function [82]. These receptor subunits adopt a modular structure featuring four distinct domains: the extracellular amino-terminal domain (ATD) which is crucial for subtype-specific receptor assembly, trafficking, and modulation; extracellular ligand-binding domain (LBD) central to agonist/competitive antagonist binding and gating, the domain forming the ion channel (TMD), and an intracellular carboxyl-terminal domain (CTD) domain involved in receptor localization and regulation [84,87]. GRI could undergo RNA editing, a mechanism that serves as a crucial modulator of the permeability properties of these receptors, encompassing the post-transcriptional modification of a particular RNA molecule that changes its coding specificity through processes different from splicing. The most prevalent form of editing is the adenosine-to-inosine (A-to-I) edition, catalyzed by adenosine deaminase acting on RNA (ADAR) I and II [88]. The translational machinery interprets these inosines as guanosines, inducing alterations in the codon of the edited nucleotide and consequent amino acid changes in the protein [89].

6. α-amino-3-hydroxy-5-methyl-4-isoxazole Propionate Receptor (GRIA)

GRIA are fast-acting Glu receptors that conduct signals at a millisecond time frame [83,90]. GRIA are heteromeric complexes composed of four subunits, namely GRIA1, GRIA2, GRIA3, and GRIA4 whose assembly generates receptors permeable to cations with distinct physiological properties dictated by their specific subunit composition [83]. GRIA1–3 mRNAs are ubiquitously expressed throughout the CNS, while GRIA4 mRNA exhibits a more restricted spatial and temporal distribution [91]. Each GRIA subunit has an extracellular N-terminal domain, the LBD features binding sites for modulators influencing receptor desensitization and deactivation, and an intracellular C-terminal tail [90].
The activation of GRIA channels allows the influx of Na+ and varying levels of Ca2+, depending on the subunits present, inducing excitatory postsynaptic currents (EPSCs) characterized by rapid kinetics and receptor desensitization, influenced by subunit composition [92,93]. Incorporation of GRIA2 limits Ca2+ permeability due to Q/R site codon editing within the pore-forming region, with all the GRIA2 subunits undergoing Q/R editing after birth, preventing divalent ion permeation [94,95]. Consequently, receptors lacking the GRIA2 subunit exhibit greater Ca2+ permeability. Alternative splicing of all GRIA subunits generates flip and flop variants with differing kinetics of desensitization, with most Flip variants exhibiting slower, less pronounced desensitization, potentially increasing vulnerability to excitotoxic injury if flop variants are decreased, as observed in amyotrophic lateral sclerosis (ALS) [96]. GRIA functions extend beyond neurons to include glial cells, in which the activation of functional GRIA regulates various cellular activities such as cell migration, morphological development, proliferation, differentiation, transcriptional regulation, survival, and ion channel regulation [97,98,99,100].

7. Kainic Acid Receptor (GRIK)

KA receptors are a type of GRI unique due to their presence at post and presynaptic terminals [101]. KA receptors also have different subunits referred to as GRIK1-5, a combination of those subunits forms tetrameric complexes [67]. GRIK comprises two subunit families, categorized by their ligand affinities: low-affinity (GRIK1-GRIK3) and high-affinity (GRIK4-GRIK5). The low-affinity subunits form homomeric or facultative heteromeric channels with both low- and high-affinity subunits. In contrast, high-affinity subunits are obligate heteromers, requiring low-affinity subunits for surface expression.
GRIK is primarily expressed in the CNS, spanning various neuronal types with substantial differences in subunit composition and splice variants across cell types and brain regions. Beyond the CNS, GRIK exhibits differential localization in various tissues, exerting significant regulatory roles in physiological activities under normal and diseased conditions. The expression levels of GRIK are intricately controlled by environmental stimulus, with specific subunits on cells acting as potent determinants of synaptic phenotypes.
The GTEx RNA-seq data highlights the notable expression of GRIK2 and GRIK5 in the cerebellar hemisphere and cerebellum, a finding consistent with supporting evidence from both in vitro and in vivo experiments [102]. Additionally, elevated GRIK5 expression is evident in diverse organs such as the testis, pituitary, colon, sigmoid, thyroid, tibial artery, and uterus, among others. Conversely, GRIK1, GRIK3, and GRIK4 predominantly exhibit expression in brain tissues.
GRIK stimulation exerts crucial modulatory roles in neurotransmission and neuronal development within the mammalian brain, being present on both pre- and post-synaptic sites of neurons [103]. While these receptors function as classical ligand-gated ion channels on postsynaptic neuronal compartments, their involvement in the non-canonical G-protein coupled metabotropic pathway on presynaptic sites allows them to finely regulate the release of excitatory and inhibitory neurotransmitters [104]. This dual modulatory capacity within neuronal circuits positions GRIK as a promising target for therapeutic interventions. Extensive research has utilized GRIKantagonists to mitigate the deleterious effects associated with various neuropathies, including epilepsy, migraine, and pain. Heteromeric GRIK composed of GRIK2 and a high-affinity GRIK subunit, such as GRIK4 or GRIK5, are enhanced by group I GRM activation through a protein kinase C (PKC)-dependent mechanism [105]. This process likely involves the phosphorylation of specific residues in the membrane-proximal C-terminus of GRIK4 or GRIK5 [106]. This regulatory mechanism imparts unique functional characteristics to heteromeric GRIK, potentially serving as a key molecular basis for group I GRM-mediated modulation of GRIK in synaptic plasticity and signal transmission [107].

8. N-methyl-D-Aspartate Receptors (GRIN)

N-methyl-D-Aspartate receptors are the third GRI subtype. These receptors are heterotetramers made up of diverse subunits encoded by three gene families GRIN1, GRIN2, and GRIN3 [108]. Functional GRIN essentially consists of GRIN1 and GRIN2 subunits [109,110]. The specific subunit composition of NMDAR exhibits variations across different synapses and developmental stages. While the GRIN1 subunit is a ubiquitously expressed essential component, GRIN2 subunits (GRIN2A-D) are encoded by distinct genes, displaying differential expression throughout the brain and during development. Notably, GRIN2A and GRIN2B expression patterns are relatively broad, with a concurrent decrease in GRIN2B and an increase in GRIN2A expression as neurons mature. GRIN2C is primarily restricted to the cerebellum and expressed later in development, whereas GRIN2D is predominantly expressed early in development, localized mainly in thalamic and hypothalamic nuclei and the brainstem [111]. The GRIN3A subunit exhibits widespread distribution early in development [112,113], while GRIN3B is primarily confined to motor neurons [114]. Endogenous GRIN typically incorporates GRIN1 and GRIN2 subunits, with GRIN3 subunits playing a modulatory role in the subpopulation of NMDAR [109].
A unique aspect of GRIN is their requisite co-agonist, glycine, in addition to Glu for activation [115]. GRIN subunits consist of a long extracellular N-terminal domain, three true transmembrane segments, a re-entrant pore loop, and an intracellular C-terminal domain of variable length. The C-terminal domain, marked by its considerable divergence among GRIN subunits, plays a pivotal role in introducing diversity to GRIN through different subunit compositions. While the N-terminal domain and extracellular loop form the ligand-binding pocket [116], the C-terminal tail regulates receptor interactions with various cytosolic proteins. These protein–protein interactions dictate the precise intracellular trafficking and localization of NMDA receptors. Additionally, different GRINR subunits are associated with distinct intracellular signaling complexes. For instance, GluN2B specifically interacts with the protein SynGAP, a Ras GTPase activating protein demonstrated to selectively inhibit NMDA-stimulated ERK signaling [117]. Furthermore, GluN2A and GluN2B exhibit differential affinities for active calcium/calmodulin-dependent protein kinase II (CaMKII), resulting in various forms of synaptic plasticity [118,119]. Lastly, the C-termini of GRIN subunits serve as substrates for post-translational modifications, such as phosphorylation. Phosphorylation is a key mechanism regulating channel function and receptor localization at synapses which influences numerous cellular processes, including protein activity, localization, and mobility [120].
In neuronal cells, GRIN channels stand out for several distinctive features, including a voltage-sensitive block by extracellular Mg2+, high permeability to Ca2+, and remarkably slow activation/deactivation kinetics [108]. The Mg2+ block serves as a molecular coincidence switch, with Mg2+ removal occurring from the channel pore during postsynaptic cell depolarization. This alleviation of the Mg2+ block, coupled with agonist binding, facilitates Ca2+ influx through the GRIN channel, subsequently modulating synaptic strength via Ca2+-activated signaling cascades [121,122,123]. Retina MCs of chick, rat, and human origin have been reported to express subtypes of NMDAR with distinctive characterization. For example, in chick retina, GRIN has been reported to have a 5-fold lower affinity for glycine and 30-fold lower affinity for D-serine when compared to the NMDAR subtypes in the brain. In addition, the GRIN subunit of the N-terminal and C-terminal splice variants also differ in retinal MCs and neurons [124,125,126].
Glial GRIN possesses a distinctive characteristic that sets them apart from neuronal counterparts—unlike neuronal GRIN, glial cells lack voltage-dependent Mg2+ block [127]. This absence of Mg2+ block is crucial for the coincidence-detecting function observed [127,128,129]. Reports from astrocytes show that NMDA-evoked currents remain unaffected by extracellular Mg2+ [130]. Intriguingly, a similar lack of prominent Mg2+ block has been noted in oligodendrocytes, where both NMDA-induced currents and NMDA-triggered i [Ca2+] transients are recorded at physiological Mg2+ concentrations [131]. The molecular basis for the low Mg2+ sensitivity of glial GRIN remains unexplained; potential explanations include specific expression of GRIN subunits or as-yet-unidentified posttranslational modifications of receptor subunits [132].

9. Metabotropic Glu Receptors (GRM)

Members of each group have a distinct pharmacological profile and can be activated selectively by agonists or allosteric modulators that have no impact on members of the other groups. Ligand binding to the VFD induces a conformational change in the GPCR, activating the G protein, comprised of α, β, and γ subunits in a heterotrimeric complex. In their inactive state, G proteins are associated with guanosine 5’-diphosphate (GDP). Activation prompts the exchange of guanosine 5’-triphosphate (GTP) for GDP within the α subunit. Subsequently, activated G protein subunits modulate various effector molecules, including enzymes, ion channels, and transcription factors. Termination of G protein activity transpires upon hydrolysis of bound GTP to GDP, leading to the reconstitution of the heterotrimer. The C-terminal region of GRM assumes a pivotal role in governing the coupling to the G protein(s), undergoes alternative splicing, and engages in interactions with intracellular signaling molecules and scaffolding proteins [73]. The recognition sequence within the C-terminal tails of both GRM1 and GRM5 facilitates binding with members of the Homer family of anchoring proteins [132,133,134,135,136,137,138]. Subsequently, Homer proteins establish connections through Shank and PSD, forming a scaffolding structure that links to IP3 receptors, GRIN, and GRIA [139]. This intricate network implies a direct connection between group I GRM and NMDAR, strategically positioning them for the regulation of NMDA receptors at hippocampal synapses [140].
A characteristic shared among G-protein-coupled receptors (GPCRs), including GRM is their capacity to regulate their cell surface density in the presence of continuous agonist stimulation [141]. The prolonged exposure of most GPCRs to their agonists induces a dampening of responsiveness, known as desensitization. Typically, this process involves a combination of mechanisms leading to receptor down-regulation, achieved through reduced receptor synthesis coupled with increased degradation of pre-existing receptors [142]. Furthermore, emerging evidence suggests a less-explored capability wherein chronic agonist exposure results in an up-regulation of the GPCR. This occurs through the recruitment of existing pools of receptors to the plasma membrane, contingent on tissue type and specific conditions [143,144]. These regulatory mechanisms, governing GRM responses to sustained high Glu levels, could play a role in either promoting or mitigating excitotoxicity [145,146].

10. Group I GRM

Group I includes GRM1 and GRM5. These receptors exhibit positive coupling to the inositol trisphosphate (IP3)-diacylglycerol (DAG) pathway. Upon ligand binding, these receptors engage positively with phospholipase C (PLC) through Gq/11 signaling [147]. Consequently, the increase in intracellular Ca2+ activates protein kinase C (PKC). The activation of PKC by group I GRM follows a canonical pathway, in which conventional PKC isotypes are activated by the rise in intracellular Ca2+ and most isotypes of PKC require both Ca2+ and DAG for optimal activity [148]. Activation of group I GRM leads to the hydrolysis of phosphatidylinositol 4,5-bisphosphate (PIP2), generating IP3 and DAG. Subsequently, IP3 induces the release of Ca2+ from intracellular stores. Glial cells exhibit the expression of all three types of inositol triphosphate receptors (InsP3Rs), potentially indicating variability across different brain regions [149]. However, a detailed mapping of InsP3Rs expression in glial cells from various origins and locations within the brain is still pending. Existing evidence suggests that the activation of InsP3Rs can elicit Ca2+ signals in diverse types of glial cells, observed in both cultured environments and in situ preparations [150].
Group I GRM possesses the characteristic membrane topology of G protein-coupled receptors (GPCRs); an extracellular N-terminus, an intracellular C-terminus (CT), three intracellular loops, and seven transmembrane domains. The intracellular domains of these receptors create a structurally spacious foundation for direct protein–protein interactions, facilitating the binding of various membrane-bound or cytoplasmic proteins [151]. Noteworthy among these binding proteins are protein kinases, including mitogen-activated protein kinases (MAPKs), Src family kinases (SFKs), Ca2+/calmodulin-dependent protein kinase II (CaMKII), and PKC [152,153]. These kinases, localized within the postsynaptic density (PSD) microdomain, bind to the intracellular domains of GRMR1/5 receptors, particularly the CT region, where they phosphorylate specific residues on the receptors [154]. Additionally, scaffolding or adaptor proteins involved in endocytosis and/or exocytosis of membrane-bound receptors engage with GRM1/5. As expected, these dynamic protein–protein interactions have functional consequences, influencing the clustering, trafficking (internalization and externalization), anchoring, and turnover of the receptors [155]. This interplay of protein–protein interactions plays a crucial role in regulating the efficacy of GRM1/5 coupling to various post-receptor signaling pathways and influencing GRM1/5-mediated synaptic plasticity.
Up to this point, several second messenger-dependent protein kinases have been associated with the desensitization of group I GRM. Activation of PKC has been identified in the agonist-induced desensitization of group I GRM [156]. Specifically, PKC-mediated phosphorylation of GRMR1α has been shown to induce receptor desensitization, as reported by Francesconi and Duvoisin in 2000 [157].
Research conducted by multiple groups has proposed that the activation and expression patterns of group I GRM may play regulatory roles in diverse aspects of neurogenesis and synaptogenesis during cortical development [158,159,160,161]. These studies, among others, indicate a strong correlation between the distribution pattern of group I GRM in specific brain regions and their distinct functional roles. Immunocytochemical and electron microscopy investigations suggest that GRM1 and GRM5 are localized in a perisynaptic manner, predominantly within postsynaptic neurons [162]. Beyond the CNS, group I GRM are also found outside the brain, where they fulfill various crucial roles. For instance, they are present in skin cells, contributing significantly to pain sensation. Additionally, these receptors are expressed in osteoblasts, hepatocytes, and heart cells [163].

11. Groups II and III mGluRs

Group II GRM is made up of GRMR2 and GRMR3 [164] while Group III GRM include GRMR4, GRMR6, GRMR7, and GRMR8 [137]. Group II and III receptors are linked to Gαi/o proteins, leading to the inhibition of adenylyl cyclase activity [165]. This inhibition results in a decrease in cAMP formation and protein kinase A activity. Group II/III receptors are either distributed both pre-synaptically and/or post-synaptically [166]. Expression analyses have revealed the presence of GRM4, GRM7, and GRM8 in both the inner nuclear layer (INL) and the ganglion cell layer, suggesting associations with multiple cell types [167]. In contrast, GRMR6 expression has been localized to the INL [168] and the outer plexiform layer (OPL) [169], where bipolar cell somata and dendrites are situated. Immunocytochemically, GRMR6 has been pinpointed to the dendritic tips of ON bipolar cells within the OPL of the rat retina, precisely where they receive input from presynaptic photoreceptor cells [169]. This localization suggests that GRMR6 plays a crucial role in mediating synaptic transmission between photoreceptors and ON bipolar cells, converting the light-induced hyperpolarization of photoreceptors into depolarization of the ON bipolar cells [170]. Glu activation of mGluR6 leads to a Gαo protein-dependent [171] closure of the transient receptor potential melanoma-related 1 (TRPM1) a non-selective cation channel presumably in a constitutively active state and cell hyperpolarization. Gαo2, an alternative splice variant of Gαo, enhances the response sensitivity [172]. A report has, however, implicated Gβγ, rather than Gαo, in the negative regulation of TRPM1 [173]. Additionally, several other proteins are implicated in modulating or scaffolding this cascade, including Gß5, RGS11, RGS7, R9AP, Ret-PCP2, nyctalopin, and potentially Ret-RGS1 [174,175].
Studies in primate retina have revealed that GRM6 aggregates approximately 400 nm away from the release site within the dendritic tips of ON cone bipolar cells, positioning these receptors at a distance comparable to that of OFF cone bipolar dendritic receptors [176,177]. The expression pattern of GRM6 is tightly regulated during development, with widespread expression throughout bipolar soma early in retinal development, followed by an increase in the OPL around the time of eye-opening (day 14), and eventual confinement to the OPL by day 28 [169]. The crucial role of mGluR6 in ON bipolar cells was first demonstrated in mice lacking GRM6, which display a lack of ERG b-wave activity, indicative of ON bipolar cell function. Consistent with this finding, spontaneous mutations in the mouse mGluR6 gene (nob3 and nob4) resulted in negative ERG waveforms and visual behavior abnormalities. Similarly, certain mutations in the human GRMR6 gene lead to autosomal recessive complete congenital stationary night blindness (arCSNB) [178].
Studies have shown that mGluR7 can reduce the activity of NMDAR through an actin-dependent process [179], while agonists targeting mGluR2 and mGluR3 can enhance NMDAR function through kinase-mediated pathways [180]. However, there is a lack of comprehensive research investigating the impact of group II and III mGluR activation on AMPAR/KAR. Recent findings have unveiled a non-standard mechanism of synaptic long-term depression triggered by mGluR3-induced internalization of AMPAR [181]. Group II agonists, Dicarboxycyclopropyl glycine (DGC-IV) and Aminopyrrolidine-2,4-dicarboxylate (APDC) have been shown to possess neuroprotective and antiepileptic effects, respectively [182,183], and reduces cell proliferation in some areas of the brain [184] and could be a therapeutic drug affecting cell apoptosis [185].

12. Distribution, Expression and Regulation of Glu Receptors in Müller Cells

Research investigations using biochemical and molecular methods revealed that Glu receptors present on cerebellum BGCs, and their adjacent Purkinje neurons, possess similar density, features, and properties. In 1992, research by Müller et al. [186] postulated that glial membranes harbor Ca2+-permeable GRIA/GRIK that, upon activation, depolarize the cells through the gating of voltage-independent channels [187] and/or electrogenic transport systems [188]. Puro [189] credited the ionotropic receptor activation to the propagation of the phosphoinositide transduction system. Notably, GRI has been identified in retinal MCs and cerebellar BGCs [125,190,191]. The GRIN expression in astrocytes in situ was initially detected by immunocytochemistry and mRNA analysis. The mRNA specific for GRIN1, GRIN2A, and GRIN2B subunits is expressed in cerebellar BGCs [190]. Glu receptor activation induces changes in the repertoire of GRIA/GRK subunits in Bergmann and Müller glial cells. Among the studied subunits, GRIA4 indicates an up-regulation in BGCs at both mRNA and protein levels [57]. Conversely, in MCs, Glu treatment led to a decrease in GRIA4 mRNA and protein expression. These effects are modulated by the group I GRM [57] GRIN1-positive cells were also found in cultured MCs isolated from the adult human retina, as confirmed by a Western blot approach [192].
Glu receptor expression is modulated through phosphorylation. In addition to serine and threonine, tyrosine represents a phosphorylation site crucial for cellular processes. Tyrosine phosphorylation is predominantly catalyzed by tyrosine kinases, encompassing both receptor and non-receptor tyrosine kinases. These enzymes play a pivotal role in modulating the expression and function of proteins by phosphorylating tyrosine residues. Within the scope of Glu receptor regulation, the Src family kinases (SFK) have emerged as a prominent subfamily of non-receptor tyrosine kinases [193]. Among the nine SFK members, five are expressed in the brain [193]. Of particular interest within this subset are Fyn (isoform 1, also known as FynB) and Src, both notably enriched at synaptic sites, suggesting their involvement in targeting local substrates to modulate synaptic transmission and plasticity [194]. Accumulating evidence underscores the roles of Fyn and Src in phosphorylating iGluRs, influencing their physiology [166,195]. Furthermore, recent findings position Fyn as a crucial SFK member in the regulation of group I GRM [193].
The expression of Glu receptors can also be downregulated through the calcium-mediated activation of calpains. Activation of these proteases may lead to the proteolysis of both GRIN and GRIA [196,197,198]. Additionally, the Ca2+-dependent activation of calcineurin and calmodulin has the potential to inhibit voltage-gated and NMDAR-gated calcium currents, respectively [199,200].

13. Glu Receptors Localization in Retinal Cells

In recent years, a multitude of studies have employed diverse immunohistochemical and electrophysiological techniques to delineate the distribution of Glu receptors in the vertebrate retina. The elaborated array of receptor subunits and transporter subtypes across various retinal cells is summarized in Table 1, reflecting the complex components accountable for retinal glutamatergic neurotransmission.

14. Glu Receptor-Mediated Signaling Events in Müller Glial Cells

Radial glial cells exhibit both short-term and long-term responses to neuronal stimulation, involving a diverse array of transcription factors. The interplay of these factors results in the differential expression of genes based on the stimulation of their neurotransmitter receptors [56]. The primary effectors of Glu receptor-mediated signaling on its target cells are Na+ and Ca2+, which are responsible for the changes in excitability and the triggering of intracellular cascades that mediate glutamatergic transmission [209]. Agonists of both GRI and GRM stimulate Ca2+-dependent transcription of various primary response genes, including those from the Fos and Jun families, in astrocytes and cells of the oligodendrocyte lineage [210]. Unlike iGluR signaling, where Glu binding directly activates ion channels, GRM signaling leads to Ca2+ influx through indirect ion channel opening and/or second messenger signaling systems [133]. In-depth exploration of intracellular signaling pathways activated by mGluR has been conducted extensively in neuronal cells, yet there is a comparatively limited understanding of glial cells [211].
In 1994, Sanchez and Ortega reported that in BGC, the activation of the AMPAR/KAR subtype increased the DNA binding activity of the activator protein 1 (AP-1) family of transcription factors in a dose- and time-dependent manner [212]. Alterations in intracellular Ca2+ concentration resulting from the activation of Glu receptors can influence gene transcription in glial cells by modulating the expression and activity of transcription factors [213]. Although the activity of Glu transporters (GLAST/EAAT1) on the membrane of the glial cells also contributes to the concurrent rise in intracellular Na+ levels, and the subsequent reverse operation of the Na+/Ca2+ exchanger (NCX), leading to an elevation in intracellular Ca2+ concentration, has emerged as a pivotal signaling mechanism in astrocytes [214]. For instance, in BGC, the downstream cascades differ; the receptor-mediated cascade leads to a PKC-mediated transcriptional response [215] and a Ca2+/calmodulin-mediated inhibition of protein synthesis elongation [216]. In contrast, Glu transporter-triggered Ca2+ influx is predominantly linked to translational control via the activation of the mechanistic target of rapamycin (mTOR) [217].
The activation of Glu-induced Ca2+ entry leads to the activation of multiple kinases including Protein Kinase C (PKC) and Ca2+/calmodulin-dependent protein kinase II (CaMKII) [218,219]. Calmodulin (CaM) is a pivotal calcium-binding protein involved in numerous cellular physiological processes [220]. Comprising two globular lobes, CaM exhibits the capability to bind two Ca2+ individually [221]. The complexity of PKC signaling is underscored by molecular cloning and sequencing, revealing a diverse family of PKC enzymes. The classical subtypes (alpha, beta I, beta II, and gamma) are activated by phorbol esters and Ca2+; the novel subtypes (delta, epsilon, eta, and theta) are also activated by phorbol esters but are Ca2+-independent; and the atypical isoforms (zeta and lambda) are not activated by phorbol esters [222]. PKCs, respond differently to cofactor dependence. Notably, PKCα and PKCε participate in the transcriptional regulation of GLAST [215]. This same group also reported that the activated GRIA subtype undergoes tyrosine phosphorylation, through a Src-dependent mechanism providing docking sites for transduction molecules like PI3K [223]. Among the signal transducers activated by phosphorylated inositides is PKC. Also, in BGC, GRIA activation is connected to the transcriptional regulation of chick kainate binding protein (chKBP) in a biphasic manner, mediated by AP-1 and Oct-2 [215,219,224,225]. In another contribution, this same group delved into the interaction between chick cerebellar GRIA and signaling proteins such as phosphorylated focal adhesion kinase (pp125FAK), PI-3K, and paxillin, both in cultured BGCs and cerebellar slices [223,226]. The dynamic interaction between transcription factors in BGC may contribute to establishing, maintaining, or inhibiting neuron-glia interactions, particularly in the context of Ca2+-permeable GRIA [99]. In a subsequent study, this same group showed that Glu and AMPA also increase the interaction of the transcription factor Ying Yang 1 (YY1) with the GLAST promoter, and overexpression of this transcription factor decreases GLAST expression [227]. These studies suggest that the effects of AMPA may depend on both AP-1 and YY1. Using the same model, Poblete-Naredo and her colleagues demonstrated that insulin increases YY1 binding to the GLAST promoter by EMSA decreasing GLAST expression [228].
Glu has been shown to modify gene expression in MCs through GRIN-mediated activation of transcription factor pathways and to evoke the release of neuroactive compounds that feedback to neurons [125,229,230]. Among these compounds, studies have identified D-serine, which is released upon Glu stimulation, mimicking the activity of glycine at the strychnine-insensitive site of GRIN, and could act as an endogenous Glu co-agonist. In the vertebrate retina, D-serine is present in MCs and astrocytes and has also been shown to contribute to the physiological activation of GRIN in retinal neurons [231]. Our findings demonstrate that selective activation of GRIN enhances [3H]-D-aspartate uptake, suggesting an increase in the abundance of GLAST/EAAT1 transporters at the plasma membrane [232]. Additionally, treatment with NMDA leads to the association of the transporter with ezrin and GFAP proteins, known to play roles in retaining GLAST at the plasma membrane [232].
Group I mGluR is known to activate MAPK, a pivotal player in protein synthesis-dependent neuronal plasticity [211,233]. The translation and transcription factors modulated by MAPK cascades following the activation of GRM have been thoroughly characterized [211]. In glial cells, particularly astrocytes, a more comprehensive analysis reveals that stimulation of MAPK and PI3K pathways via GRM3 enhances the production of neurotrophic factors, promoting neuroprotection against various toxic insults [234,235,236]. In Group III GRM, the activation of GRM4 receptors in cultured rat neural stem cells leads to the inhibition of JNK and p38 mitogen-activated protein kinase. This downregulates the expression of procaspase-8/9/3 and reverses the Bcl-2/Bax balance, ultimately preventing H2O2-mediated cell death [237]. Additionally, a protective role for mGluR7 has recently been discovered in glial cells, involving the activation of PI3K/Akt and MAPK-ERK1/2 pathways [238]. Activation of group II GRM up-regulates GLAST mRNA and protein levels [239,240] via the extracellular signal-regulated kinase (ERK)/PI3K/NF-κB pathway [241]. A summary of these findings is presented in Figure 3.

15. Impact of Glu Receptor Signaling in Excitotoxicity and Retinal Degeneration

The visual system is susceptible to a myriad of pathophysiological conditions collectively referred to as retinal degenerative diseases characterized by distinct clinical pathology and features [242,243,244]. The progressive accumulation of Glu in the extracellular space and over-activation of neuronal Glu receptors has been linked as one of the main causes of retinal diseases. Glu possesses potent excitotoxic properties towards neuronal cells and exposure to micromolar concentrations can lead to both immediate and delayed cell death [245,246]. In cultured retinal neurons, the activation of GRIN and the subsequent influx of Ca2+ are considered the primary mechanisms underlying Glu neurotoxicity [177,247]. Excessive extracellular Glu concentration in RGCs leads to uncontrolled continuous depolarization, termed excitotoxicity [248]. Excitotoxicity is characterized by neuronal death due to prolonged Glu exposure and GRIA and GRIN activation not only at the synaptic cleft, but importantly, the activation of extra synaptic GRIN receptors, with a differential subunit composition (enrichment of highly Ca2+ permeable GRIN2B subunits) and the consequent excessive Ca2+ influx that activates enzymes responsible for the degradation of proteins, membranes, and nucleic acids [249], [250]. GRMs contribute as well, to the increase in intracellular Ca2+ concentrations [251]. In the brain, the activation of GRIA and GRIN are involved in synaptic plasticity, culminating in the induction of long-term potentiation (LTP), a sustained enhancement in signal transmission [252,253]. These receptors facilitate the influx of Ca2+ into the cell, triggering various intracellular signaling pathways, receptor trafficking, gene expression, and the generation of LTP.
Glu receptor subunit composition and expression could determine the influx of ions, particularly the GRIA2 subunit with widespread expression patterns [254]. While most neurons containing GRIA express GRIA2, there are variations in the expression levels. In numerous brain regions, approximately 8–15% of neurons express Ca2+-permeable GRIA lacking GRIA2, and these specific neuronal subpopulations are selectively eliminated in a Ca2+-dependent manner when exposed to either AMPA or KA [66,243]. The presence of low but persistent permeability to Ca2+ in heterogeneous cell populations might elucidate why mammalian neurons are highly susceptible to GRIA-mediated damage, such as hippocampal granule and pyramidal neurons [255] as well as cortical neurons [256], given the fact that these cells still express GRIA2. However, it remains uncertain whether low Ca2+ permeability is the sole factor responsible for GRIA-mediated excitotoxic cell death, although evidence from experiments involving mice lacking a functional GRIA2 gene suggests that increased Ca2+ influx in CA1 neurons does not lead to neuropathological lesions indicative of excitotoxicity [257,258]. Furthermore, in MCs, activation of nuclear factor kappa B (NF-kB) leads to increased surface levels of Ca2+-permeable GRIA in RGCs, triggering cell death [259].
The intracellular accumulation of Ca2+ serves as the trigger for activating protein kinases and other downstream Ca2+-dependent enzymes, ultimately leading to cell death through molecular modifications such as cellular membrane damage, the release of reactive oxygen species (ROS), mitochondrial dysfunction, upregulation of neuronal nitric oxide synthase (nNOS), endoplasmic reticulum (ER) stress, and release of lysosomal enzymes, culminating in excitotoxic, oxidative, and apoptotic stress [260,261,262]. Ca2+/calmodulin-dependent protein kinase II (CaMKII) is a protein kinase subject to regulation by the Ca2+/calmodulin complex [263]. In certain neuronal disorders such as stroke, epilepsy, and glaucoma, increased Ca2+ levels may dysregulate CaMKII activity [264,265,266]. In preventing neuronal damage both in vitro and in vivo, CaMKII inhibition before excitotoxic injury emerges as a potential therapeutic strategy [267]. This emphasizes the significance of understanding the role of CaM and CaMKII in the context of Ca2+ regulation, offering valuable insights into the pathophysiology of various neurological conditions [268].
GRIA featuring Q/R-unedited GRIA2 subunits also exhibit permeability to zinc, potentially implicating zinc in processes related to differentiation [269,270]. Studies have demonstrated that astrocytes with mutant superoxide dismutase type 1 (SOD1) release factors that decrease the expression of this specific subunit in motor neurons. This, in turn, results in AMPAR-mediated excitotoxicity and eventual cell death [271,272]. There is experimental evidence indicating that the excessive activation of Ca2+-permeable GRIA containing GRIA4 is linked to a death signaling pathway, which is influenced, at least in part, by the AP-1 transcription factor [273]. Furthermore, in the rat retina, GRM activation has been shown to mitigate the detrimental impact of NMDA [274]. Additionally, neuronal injuries give rise to adaptive alterations in GRM expression reminiscent of developmental patterns. During development, RGCs express transcripts for various types of mGluR, some of which decrease in expression during adulthood. However, following neuronal injury, this developmental downregulation appears to be reversed [177].
The impact of elevated levels of extracellular Glu is exacerbated when the activity of the Glu uptake/transporter system is compromised [275,276,277,278]. Dysfunction in the Glu transport into MCs contributes to an increase in extracellular Glu, reaching excitotoxic levels. Research evidence has demonstrated the vulnerability of astrocytic Gln synthetase to inactivation induced by reactive oxygen species (ROS), disrupting the entire Glu/Gln cycle and contributing to an elevation in extracellular Glu concentration, thereby promoting excitotoxicity [279]. Furthermore, the presence of ROS has been observed to diminish Glu transporter activity, hindering the synaptic clearance of Glu and further amplifying the rise in extracellular Glu concentration [53]. Oxygen deprivation can also trigger excitotoxicity through a rather distinct mechanism. Directly, hypoxic-ischemic conditions stimulate Glu release, escalating the extracellular Glu concentration to neurotoxic levels in a straightforward manner [280]. Additionally, oxygen deprivation induces energetic stress by disrupting mitochondrial oxidative phosphorylation, thereby impeding ATP production [281]. This disruption results in intracellular ATP depletion, preventing the reuptake of Glu resulting in an excitotoxic Glu extracellular concentration [282]. The main utilizers of ATP in the brain are ion pumps responsible for sustaining ion gradients crucial for neuronal signaling and communication [283,284,285]. The effect of excessive Glu release leads to the uncontrolled stimulation of Glu receptors, which leads to the massive influx of Ca2+ into postsynaptic neurons due to low ATP levels, hence impairing ion pump function [286,287].

16. Role of Müller Glial Cells in Retinal Diseases Prevention

In retinal degeneration, MC physiology has been studied to detect early signs of retinal neuronal death. Studies have shown that early or detectable stages of retinal degeneration are characterized by alterations of important features of MCs such as an increased expression of glial fibrillary acidic protein (GFAP), cellular retinaldehyde-binding protein (CRALBP) and Gln synthetase, undergoing varying degrees of loss [288]. In the later stages of retinal diseases, the metabolic profile and protein expression of Müller cells exhibit a distinctive “chaotic” pattern [289].
The inhibition of Gln synthetase in MCs through pharmacological blockade leads to a reduction in the Glu levels within bipolar and ganglion cells, resulting in functional blindness [290]. Gln synthetase downregulation contributes to neuronal dysfunction and exacerbates Glu toxicity in diverse retinopathies [291]. Glu accumulation inhibits cystine uptake, reinforcing the event by depleting cysteine and glutathione’s reducing potential. Glu-induced nitric oxide (NO) generation and subsequent interactions with oxygen radicals contribute to delayed death of retinal neurons in retinal ischemia, further accelerating RGC death.
Understanding the cellular mechanisms involved in the impairment of glial Glu recycling may help to find novel targets for developing neuroprotective agents. An increase in Gln synthetase in MCs may represent an attractive approach that has been shown to protect against neuronal degeneration in the injured retinal tissue [292]. The decrease in uptake might be caused by the downregulation/inactivation of Gln synthetase, by mitochondria-derived free radicals, which inactivate the transporter molecules, as well as by cell depolarization resulting from high extracellular K+, energy failure due to mitochondrial dysfunction, downregulation/inactivation of K+ channels, and inhibition of Na*/K+ ATPase and Glt-1 through inflammatory lipid mediators [291].
Prolonged exposure to Glu can induce alterations in the morphological features of glial cells without necessarily leading to cell death. When retinal MCs are exposed to Glu, the cytoskeleton undergoes a collapse, resulting in a transition from an epithelioid to a retracted morphology [187]. These morphological modifications might potentially impact the ability of MCs to regulate extracellular K+ levels released during neuronal activity [79] and the expression of neurotransmitter receptors and transporters situated in the membrane.
Transient retinal ischemia or diabetes does not significantly alter GLAST expression or its evoked membrane currents, it reduces the efficiency of Glu transport into MCs. This leads to a high Glu influx into photoreceptor, bipolar, and ganglion cells under these conditions. In patients with glaucoma, a downregulation of GLAST (but not Glt-1) has been observed [276]. Increased intraocular pressure in experimental glaucoma results in a failure of GLAST activity, leading to decreased Glu accumulation in MCs and significant Glu uptake by RGCs, coinciding with excitotoxic damage to the retina [243]. The elevated intraocular pressure causes inner retinal hypoxia, triggering the formation of free radicals, and disrupting Glu transport in MCs. Similar dysfunction in electrogenic Glu transport caused by oxidative stress is observed in experimental diabetes [293].
This malfunction in Glu transport into MCs, induced by free radicals from mitochondria, may explain retinal ganglion cell death in Leber hereditary optic neuropathy [294]. MCs from patients with retinopathies display increased density of Glu transporter currents, and experimental retinal detachment shows an increase in GLAST labeling. However, under these conditions, MCs depolarize due to functional inactivation or downregulation of inwardly rectifying K+ channels (Kir) [295]. Various retinal diseases associated with a breakdown of blood-retinal barriers result in serum leakage into the retina, impacting Glu transport efficiency. Clinical observations link hemorrhages at vascular leakage sites with a greater reduction in retinal function, possibly linked to the decreased efficiency of Glu transport into MCs [296]. The depolarization of MCs may be strong enough to reverse the operation mode of Glt-1 resulting in an efflux of Glu that contributes to excitotoxic neuronal damage. A significant portion of glial Glu might also be released through the cystine-glutamate antiporter and vesicular exocytosis [69].
Above we describe the signaling pathways and how these could be activated by Glu receptors. These pathways have an important role in different organisms. Diverse studies have demonstrated the activation of phosphoinositide (PI) hydrolysis in MCs which activate the PKC pathway [230]. The PKC family might be involved in regulating synaptic actions in glutamatergic systems such as blood-brain barrier homeostasis, apoptotic cycle, immune regulation, signal transduction, and transmission which allows the development and differentiation of tissues and cells; those activities give these enzymes an important role in physiology [297,298]. Activation of PKC is also involved in the PLC pathway which is characterized to activate and generate the second messengers DAG and PIP2. Generally, PLC allows lipidic hydrolysis and second messengers generation, and that leads to cell regulation by proliferation and migration; PLC regulates immune responses, apoptotic cycle, and neuronal signaling [299]. Due to their involvement in these important processes, dysfunction in the pathway most possibly leads to disease. In retina illnesses such as Diabetic retinopathy, resulting from damaged retinal blood vessels, it could lead to permanent blindness. An increase in PKC activity has been reported in the generation of new blood vessels, a process known as neovascularization [300,301].
Several diseases characterized by reduced Glu transporter expression are linked to alterations in DNA methylation patterns. Recently, our group reported the effect of Glu on global 5-methylcytosine (5-mc) content in radial glial cells, this study implicates dynamic DNA methylation/demethylation in gene expression regulation of glial Glu transporters [302]. Retinal disorders such as age-related macular degeneration and diabetic retinopathy are linked to dynamic DNA methylation [303]. For instance, hypermethylation of the Glt-1 promoter is observed in brain tumors [304]. Aberrant methylation control underlies Rett syndrome, a neurodevelopmental disorder resulting from mutations in the DNMT methyl-CpG-binding protein 2 (MeCP2), resulting in dysregulation of both GLAST and Glt-1 [305,306,307].
An essential progression from these molecular investigations and a future research challenge, involves identifying downstream genes regulated by transcription factors associated with Glu-receptor-mediated signaling in glia. For instance, some clinical studies reported that many Glu agonists and antagonists exhibit neuroprotective effects in retinal neuronal cells, and t-ACPD, a mGluR Group I and II agonist, possesses neuroprotective effects against ischemic damage [308]. Also, the mGluR1 antagonist, CPCCOEt could possess neuroprotective abilities against excitotoxic-induced death [309], similarly, an agonist of mGluR group II, like LY354740 has a neuroprotective effect that is enhanced by glial cells [310], in addition to that, in the neuroprotective effect of Group II, it is believed that transforming growth factor- β (TGFβ) is the main intermediary [311]. Likewise, a GRI antagonist, such as ifenprodil, could produce, like the other agonists and antagonists, a neuroprotective effect without modifying the synaptic transmission [312]. The Group II agonist LY354740 reverses RGC hyperexcitability and increases the expression level of Brain-derived neurotrophic factor (BDNF), thus providing a neuroprotective effect [313].

17. Perspectives

The evidence reviewed above strongly suggests a complex functional interaction between retinal neurons and MCs hence, influencing their information processing and behavioral output. Glu receptors modulating glutamatergic neurotransmission are expressed on both neuronal and glial cells, this similarity makes it complicated to modulate their downstream activities as this would lead to various side effects that might complicate the assessment of therapeutic efficacy. However, a promising alternative could be exploring glial-cell-specific downstream molecular targets which present an avenue to develop optimized therapeutics. Current evidence is limited to in vitro and brain slice experiments, which limit our understanding of the functional role of all these components in vivo.
This review highlights the relevance of Glu receptor activation on glial-neuronal communication in the retina. Arising questions are still to be discussed. What influences the differential expression patterns of Glu receptors? How does Glu receptor activation affect visual functions at molecular and physiological levels? What are the different regulatory mechanisms through Glu receptor signaling involved in the expression of Glu transporters? How do epigenetic modifications, such as DNA methylation and histone modifications, regulate the expression of Glu receptors and transporters? What signaling pathways and transcription factors are involved in the regulation of Glu transporters, and how do they contribute to neurological disorders?

18. Conclusions

We focused on Glu receptor-mediated signaling in MCs due to their significant role in modulating glutamatergic transmission in the retina. For many years, there has been a paucity of adequate information on the role of MC signaling in ocular diseases, but recent research has made it clear that glial cells are indispensable for understanding the mechanism of retinal diseases. In the future, a large effort will be required for the detailed localization of the different classes of GRM and GRI and their various subunits on the MCs, because the functional properties of receptors depend on the subunit composition, it will be especially important to determine which receptor subunits are expressed at the synapse during the light/dark cycle. Still, further research is needed to bridge the substantial gaps in our understanding of mechanisms and physiological relevance of glial-neuron glutamatergic interactions, in particular, the impact of glial Glu receptor activation and its emerging roles in retinal degeneration and diseases.

Author Contributions

B.O., Y.M.-S., T.S., L.C.H.-K. and T.N.O.-B. gathered the information, wrote the first draft and made the Figures, M.-P.F.-S. and A.O. conceptualized the work, edited the manuscript and obtained the funding. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Conahcyt-Mexico, grant number CF-I-2023-935 to A.O.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Acknowledgments

L.C.H.-K. acknowledges Posgrado en Ciencias Biológicas, Universidad Autónoma de Tlaxcala, México.

Conflicts of Interest

The authors declare no conflicts of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

References

  1. Masland, R.H. The fundamental plan of the retina. Nat. Neurosci. 2001, 4, 877–886. [Google Scholar] [CrossRef]
  2. Mannu, G.S. Retinal phototransduction. Neurosciences 2014, 19, 275–280. [Google Scholar] [PubMed]
  3. Ptito, M.; Bleau, M.; Bouskila, J. The Retina: A Window into the Brain. Cells 2021, 10, 3269. [Google Scholar] [CrossRef]
  4. Rodieck, R.W. The Vertebrate Retina: Principles of Structure and Function; W.H. Freeman & Co Ltd.: San Francisco, CA, USA, 1973. [Google Scholar]
  5. Diamond, J.S. Inhibitory Interneurons in the Retina: Types, Circuitry, and Function. Annu. Rev. Vis. Sci. 2017, 3, 1–24. [Google Scholar] [CrossRef]
  6. Baude, A.; Nusser, Z.; Roberts, J.D.B.; Mulvihill, E.; Mcllhinney, R.A.J.; Somogyi, P. The metabotropic glutamate receptor (mGluRlα) is concentrated at perisynaptic membrane of neuronal subpopulations as detected by immunogold reaction. Neuron 1993, 11, 771–787. [Google Scholar] [CrossRef] [PubMed]
  7. Shigemoto, R.; Kulik, A.; Roberts, J.D.B.; Ohishi, H.; Nusser, Z.; Kaneko, T.; Somogyi, P. Target-cell-specific concentration of a metabotropic glutamate receptor in the presynaptic active zone. Nature 1996, 381, 523–525. [Google Scholar] [CrossRef] [PubMed]
  8. Franze, K.; Grosche, J.; Skatchkov, S.N.; Schinkinger, S.; Foja, C.; Schild, D.; Uckermann, O.; Travis, K.; Reichenbach, A.; Guck, J. Müller cells are living optical fibers in the vertebrate retina. Proc. Natl. Acad. Sci. USA 2007, 104, 8287–8292. [Google Scholar] [CrossRef] [PubMed]
  9. Reichenbach, A.; Bringmann, A. New functions of Müller cells. Glia 2013, 61, 651–678. [Google Scholar] [CrossRef]
  10. Bringmann, A.; Pannicke, T.; Grosche, J.; Francke, M.; Wiedemann, P.; Skatchkov, S.; Osborne, N.; Reichenbach, A. Müller cells in the healthy and diseased retina. Prog. Retin. Eye Res. 2006, 25, 397–424. [Google Scholar] [CrossRef] [PubMed]
  11. Reichenbach, A.; Faude, F.; Enzmann, V.; Bringmann, A.; Pannicke, T.; Francke, M.; Biedermann, B.; Kuhrt, H.; Stolzenburg, J.-U.; Skatchkov, S.N.; et al. The Müller (Glial) Cell in normal and diseased retina: A case for single-cell electrophysiology. Ophthalmic Res. 1997, 29, 326–340. [Google Scholar] [CrossRef]
  12. Tsunoda, S. Organization of Photoreceptor Signaling Complexes. In Handbook of Cell Signaling, 2nd ed.; Academic Press: Cambridge, MA, USA, 2010; Volume 1, pp. 373–377. [Google Scholar] [CrossRef]
  13. Yin, L.; Smith, R.G.; Sterling, P.; Brainard, D.H. Chromatic properties of horizontal and ganglion cell responses follow a dual gradient in cone opsin expression. J. Neurosci. 2006, 26, 12351–12361. [Google Scholar] [CrossRef] [PubMed]
  14. Borghuis, B.G.; Ratliff, C.P.; Smith, R.G. Impact of light-adaptive mechanisms on mammalian retinal visual encoding at high light levels. J. Neurophysiol. 2018, 119, 1437–1449. [Google Scholar] [CrossRef]
  15. Moini, J.; LaGalbo, A.; Ahangari, R. The visual system. In Foundations of the Mind, Brain, and Behavioral Relationships: Understanding Physiological Psychology; Elsevier: London, UK, 2023; Chapter 8; pp. 125–142. [Google Scholar]
  16. Remington, L.A.; Goodwin, D. Retina. In Clinical Anatomy and Physiology of the Visual System, 4th ed.; Elsevier: St. Louis, MO, USA, 2022; Chapter 8; pp. 111–139. [Google Scholar]
  17. Jo, A.; Deniz, S.; Xu, J.; Duvoisin, R.M.; DeVries, S.H.; Zhu, Y. A sign-inverted receptive field of inhibitory interneurons provides a pathway for ON-OFF interactions in the retina. Nat. Commun. 2023, 14, 5937. [Google Scholar] [CrossRef] [PubMed]
  18. Dieck, S.T.; Brandstätter, J.H. Ribbon synapses of the retina. Cell Tissue Res. 2006, 326, 339–346. [Google Scholar] [CrossRef]
  19. Reichenbach, A.; Henke, A.; Eberhardt, W.; Reichelt, W.; Dettmer, D. K+ ion regulation in retina. Can. J. Physiol. Pharmacol. 1992, 70, S239–S247. [Google Scholar] [CrossRef] [PubMed]
  20. Fykse, E.M.; Fonnum, F. Amino acid neurotransmission: Dynamics of vesicular uptake. Neurochem. Res. 1996, 21, 1053–1060. [Google Scholar] [CrossRef] [PubMed]
  21. Hurley, J.B. Phototransduction. In Encyclopedia of Neuroscience; Academic Press: Cambridge, MA, USA, 2009; pp. 687–692. [Google Scholar] [CrossRef]
  22. Thoreson, W.B.; Witkovsky, P. Glutamate receptors and circuits in the vertebrate retina. Prog. Retin. Eye Res. 1999, 18, 765–810. [Google Scholar] [CrossRef] [PubMed]
  23. Massey, S.C.; Miller, R.F. N-methyl-D-aspartate receptors of ganglion cells in rabbit retina. J. Neurophysiol. 1990, 63, 16–30. [Google Scholar] [CrossRef] [PubMed]
  24. Massey, S.C. Cell types using glutamate as a neurotransmitter in the vertebrate retina. Prog. Retin. Res. 1990, 9, 399–425. [Google Scholar] [CrossRef]
  25. Naito, S.; Ueda, T. Adenosine triphosphate-dependent uptake of glutamate into protein I-associated synaptic vesicles. J. Biol. Chem. 1983, 258, 696–699. [Google Scholar] [CrossRef] [PubMed]
  26. Tabb, J.S.; Ueda, T. Phylogenetic studies on the synaptic vesicle glutamate transport system. J. Neurosci. 1991, 11, 1822–1828. [Google Scholar] [CrossRef] [PubMed]
  27. DeVries, S.H. Bipolar cells use kainate and AMPA receptors to filter visual information into separate channels. Neuron 2000, 28, 847–856. [Google Scholar] [CrossRef] [PubMed]
  28. Borghuis, B.G.; Looger, L.L.; Tomita, S.; Demb, J.B. Kainate Receptors Mediate Signaling in Both Transient and Sustained OFF Bipolar Cell Pathways in Mouse Retina. J. Neurosci. 2014, 34, 6128–6139. [Google Scholar] [CrossRef] [PubMed]
  29. Ichinose, T.; Habib, S. On and off signaling pathways in the retina and the visual system. Front. Ophthalmol. 2022, 2, 989002. [Google Scholar] [CrossRef] [PubMed]
  30. Puthussery, T.; Percival, K.A.; Venkataramani, S.; Gayet-Primo, J.; Grünert, U.; Taylor, W.R. Kainate receptors mediate synaptic input to transient and sustained OFF visual pathways in primate retina. J. Neurosci. 2014, 34, 7611–7621. [Google Scholar] [CrossRef]
  31. Nakajima, Y.; Iwakabe, H.; Akazawa, C.; Nawa, H.; Shigemoto, R.; Mizuno, N.; Nakanishi, S. Molecular characterization of a novel retinal metabotropic glutamate receptor mGluR6 with a high agonist selectivity for L-2-amino-4-phosphonobutyrate. J. Biol. Chem. 1993, 268, 11868–11873. [Google Scholar] [CrossRef]
  32. Ichinose, T.; Hellmer, C.B. Differential signalling and glutamate receptor compositions in the OFF bipolar cell types in the mouse retina. J. Physiol. 2016, 594, 883–894. [Google Scholar] [CrossRef] [PubMed]
  33. Popova, E. ON-OFF Interactions in the Retina: Role of Glycine and GABA. Curr. Neuropharmacol. 2015, 12, 509–526. [Google Scholar] [CrossRef] [PubMed]
  34. Matthews, G.; Fuchs, P. The diverse roles of ribbon synapses in sensory neurotransmission. Nat. Rev. Neurosci. 2010, 11, 812–822. [Google Scholar] [CrossRef] [PubMed]
  35. Aizenman, E.; Frosch, M.P.; Lipton, S.A. Responses mediated by excitatory amino acid receptors in solitary retinal ganglion cells from rat. J. Physiol. 1988, 396, 75–91. [Google Scholar] [CrossRef] [PubMed]
  36. Sasaki, T.; Kaneko, A. L-Glutamate-induced responses in OFF-type bipolar cells of the cat retina. Vis. Res. 1996, 36, 787–795. [Google Scholar] [CrossRef] [PubMed]
  37. Olivares-Bañuelos, T.N.; Chí-Castañeda, D.; Ortega, A. Glutamate transporters: Gene expression regulation and signaling properties. Neuropharmacology 2019, 161, 107550. [Google Scholar] [CrossRef]
  38. Kanai, Y.; Hediger, M.A. Primary structure and functional characterization of a high-affinity glutamate transporter. Nature 1992, 360, 467–471. [Google Scholar] [CrossRef] [PubMed]
  39. Pines, G.; Danbolt, N.C.; Bjørås, M.; Zhang, Y.; Bendahan, A.; Eide, L.; Koepsell, H.; Storm-Mathisen, J.; Seeberg, E.; Kanner, B.I. Cloning and expression of a rat brain L-glutamate transporter. Nature 1992, 360, 464–467. [Google Scholar] [CrossRef]
  40. Schultz, K.; Stell, W.K. Immunocytochemical localization of the high-affinity glutamate transporter, EAAC1, in the retina of representative vertebrate species. Neurosci. Lett. 1996, 211, 191–194. [Google Scholar] [CrossRef] [PubMed]
  41. Rodríguez-Campuzano, A.G.; Ortega, A. Glutamate transporters: Critical components of glutamatergic transmission. Neuropharmacology 2021, 192, 108602. [Google Scholar] [CrossRef]
  42. Ward, M.M.; Jobling, A.I.; Puthussery, T.; Foster, L.E.; Fletcher, E.L. Localization and expression of the glutamate transporter, excitatory amino acid transporter 4, within astrocytes of the rat retina. Cell Tissue Res. 2004, 315, 305–310. [Google Scholar] [CrossRef]
  43. Fyk-Kolodziej, B.; Qin, P.; Dzhagaryan, A.; Pourcho, R.G. Differential cellular and subcellular distribution of glutamate transporters in the cat retina. Vis. Neurosci. 2004, 21, 551–565. [Google Scholar] [CrossRef] [PubMed]
  44. Arriza, J.L.; Eliasof, S.; Kavanaugh, M.P.; Amara, S.G. Excitatory amino acid transporter 5, a retinal glutamate transporter coupled to a chloride conductance. Proc. Natl. Acad. Sci. USA 1997, 94, 4155–4160. [Google Scholar] [CrossRef]
  45. Schneider, N.; Cordeiro, S.; Machtens, J.P.; Braams, S.; Rauen, T.; Fahlke, C. Functional properties of the retinal glutamate transporters GLT-1c and EAAT5. J. Biol. Chem. 2014, 289, 1815–1824. [Google Scholar] [CrossRef] [PubMed]
  46. Brew, H.; Attwell, D. Electrogenic glutamate uptake is a major current carrier in the membrane of axolotl retinal glial cells. Nature 1987, 327, 707–709. [Google Scholar] [CrossRef]
  47. Eliasof, S.; Werblin, F. Characterization of the glutamate transporter in retinal cones of the tiger salamander. J. Neurosci. 1993, 13, 402–411. [Google Scholar] [CrossRef] [PubMed]
  48. Lewerenz, J.; Maher, P. Chronic glutamate toxicity in neurodegenerative diseases—What is the evidence? Front. Neurosci. 2015, 9, 469. [Google Scholar] [CrossRef]
  49. Shigeri, Y.; Seal, R.P.; Shimamoto, K. Molecular pharmacology of glutamate transporters, EAATs and VGLUTs. Brain Res. Rev. 2004, 45, 250–265. [Google Scholar] [CrossRef]
  50. Hayashi, M.K. Structure-Function Relationship of Transporters in the Glutamate-Glutamine Cycle of the Central Nervous System. Int. J. Mol. Sci. 2018, 19, 1177. [Google Scholar] [CrossRef] [PubMed]
  51. Sarthy, V.P.; Pignataro, L.; Pannicke, T.; Weick, M.; Reichenbach, A.; Harada, T.; Tanaka, K.; Marc, R. Glutamate transport by retinal Müller cells in glutamate/aspartate transporter-knockout mice. Glia 2005, 49, 184–196. [Google Scholar] [CrossRef] [PubMed]
  52. Magi, S.; Piccirillo, S.; Amoroso, S.; Lariccia, V. Excitatory amino acid transporters (Eaats): Glutamate transport and beyond. Int. J. Mol. Sci. 2019, 20, 5674. [Google Scholar] [CrossRef] [PubMed]
  53. Anderson, C.M.; Swanson, R.A. Astrocyte Glutamate Transport: Review of Properties, Regulation, and Physiological Functions. Glia 2000, 32, 1–14. [Google Scholar] [CrossRef]
  54. Vandenberg, R.J.; Ryan, R.M. Mechanisms of glutamate transport. Physiol. Rev. 2013, 93, 1621–1657. [Google Scholar] [CrossRef] [PubMed]
  55. Danbolt, N.C. Glutamate uptake. Prog. Neurobiol. 2001, 65, 1–105. [Google Scholar] [CrossRef]
  56. Cameron, R.S.; Rakic, P. Glial cell lineage in the cerebral cortex: A review and synthesis. Glia 1991, 4, 124–137. [Google Scholar] [CrossRef] [PubMed]
  57. López, T.; López-Colomé, A.M.; Ortega, A. Changes in GluR4 expression induced by metabotropic receptor activation in radial glia cultures. Mol. Brain Res. 1998, 58, 40–46. [Google Scholar] [CrossRef]
  58. Kettenmann, H. Beyond the neuronal circuitry: Introduction. Trends Neurosci. 1996, 19, 305–306. [Google Scholar] [CrossRef] [PubMed]
  59. Hogan, M.J.; Alvarado, J.A.; Weddell, J.E. Histology of the Human Eye: An Atlas and Textbook, 1st ed.; Saunders: St. Louis, MO, USA, 1971. [Google Scholar]
  60. Vecino, E.; Rodriguez, F.D.; Ruzafa, N.; Pereiro, X.; Sharma, S.C. Glia-neuron interactions in the mammalian retina. Prog. Retin. Eye Res. 2016, 51, 1–40. [Google Scholar] [CrossRef]
  61. Jeon, C.J.; Strettoi, E.; Masland, R.H. The major cell populations of the mouse retina. J. Neurosci. 1998, 18, 8936–8946. [Google Scholar] [CrossRef] [PubMed]
  62. Grosche, J.; Härtig, W.; Reichenbach, A. Expression of glial fibrillary acidic protein (GFAP), glutamine synthetase (GS), and Bcl-2 protooncogene protein by Müller (glial) cells in retinal light damage of rats. Neurosci. Lett. 1995, 185, 119–122. [Google Scholar] [CrossRef]
  63. Riepe, R.E.; Norenburg, M.D. Müller cell localisation of glutamine synthetase in rat retina. Nature 1977, 268, 654–655. [Google Scholar] [CrossRef] [PubMed]
  64. Xu, Q.A.; Boerkoel, P.; Hirsch-Reinshagen, V.; Mackenzie, I.R.; Hsiung, G.-Y.R.; Charm, G.; To, E.F.; Liu, A.Q.; Schwab, K.; Jiang, K.; et al. Müller cell degeneration and microglial dysfunction in the Alzheimer’s retina. Acta Neuropathol. Commun. 2022, 10, 145. [Google Scholar] [CrossRef] [PubMed]
  65. Malchow, R.P.; Qian, H.; Ripps, H. gamma-Aminobutyric acid (GABA)-induced currents of skate Muller (glial) cells are mediated by neuronal-like GABAA receptors. Proc. Natl. Acad. Sci. USA 1989, 86, 4326–4330. [Google Scholar] [CrossRef] [PubMed]
  66. Wakakura, M.; Yamamoto, N. Cytosolic calcium transient increase through the AMPA/kainate receptor in cultured Müller cells. Vis. Res. 1994, 34, 1105–1109. [Google Scholar] [CrossRef] [PubMed]
  67. Boccuni, I.; Fairless, R. Retinal Glutamate Neurotransmission: From Physiology to Pathophysiological Mechanisms of Retinal Ganglion Cell Degeneration. Life 2022, 12, 638. [Google Scholar] [CrossRef]
  68. Erecińska, M.; Silver, I.A. Metabolism and role of glutamate in mammalian brain. Prog. Neurobiol. 1990, 35, 245–296. [Google Scholar] [CrossRef] [PubMed]
  69. Bringmann, A.; Pannicke, T.; Biedermann, B.; Francke, M.; Iandiev, I.; Grosche, J.; Wiedemann, P.; Albrecht, J.; Reichenbach, A. Role of retinal glial cells in neurotransmitter uptake and metabolism. Neurochem. Int. 2009, 54, 143–160. [Google Scholar] [CrossRef] [PubMed]
  70. Poitry-Yamate, C.; Gianoncelli, A.; Kaulich, B.; Kourousias, G.; Magill, A.W.; Lepore, M.; Gajdosik, V.; Gruetter, R. Feasibility of direct mapping of cerebral fluorodeoxy-D-glucose metabolism in situ at subcellular resolution using soft X-ray fluorescence. J. Neurosci. Res. 2013, 91, 1050–1058. [Google Scholar] [CrossRef] [PubMed]
  71. López-Bayghen, E.; Ortega, A. Glial glutamate transporters: New actors in brain signaling. IUBMB Life 2011, 63, 816–823. [Google Scholar] [CrossRef]
  72. Newman, E.; Reichenbach, A. The Müller cell: A functional element of the retina. Trends Neurosci. 1996, 19, 307–312. [Google Scholar] [CrossRef] [PubMed]
  73. Martinez-Lozada, Z.; Guillem, A.M.; Robinson, M.B. Transcriptional Regulation of Glutamate Transporters: From Extracellular Signals to Transcription Factors. In Advances in Pharmacology; Academic Press: Cambridge, MA, USA, 2016; Volume 76, pp. 103–145. [Google Scholar] [CrossRef]
  74. Martínez-Lozada, Z.; Ortega, A. Glutamatergic transmission: A matter of three. Neural Plast. 2015, 2015, 787396. [Google Scholar] [CrossRef]
  75. Flores-Méndez, M.; Mendez-Flores, O.G.; Ortega, A. Glia plasma membrane transporters: Key players in glutamatergic neurotransmission. Neurochem. Int. 2016, 98, 46–55. [Google Scholar] [CrossRef] [PubMed]
  76. Linser, P.; Moscona, A.A. Induction of glutamine synthetase in embryonic neural retina: Localization in Müller fibers and dependence on cell interactions. Proc. Natl. Acad. Sci. USA 1979, 76, 6476–6480. [Google Scholar] [CrossRef]
  77. Lewis, G.P.; Erickson, P.A.; Kaska, D.D.; Fisher, S.K. An immunocytochemical comparison of Müller cells and astrocytes in the cat retina. Exp. Eye Res. 1988, 47, 839–853. [Google Scholar] [CrossRef]
  78. López-Bayghen, E.; Rosas, S.; Castelán, F.; Ortega, A. Cerebellar Bergmann glia: An important model to study neuron-glia interactions. Neuron Glia Biol. 2007, 3, 155–167. [Google Scholar] [CrossRef]
  79. Heidinger, V.R.; Dreyfus, H.; Sahel, J.; Christen, Y.; Hicks, D. Excitotoxic Damage of Retinal Glial Cells Depends Upon Normal Neuron-Glial Interactions. Glia 1998, 23, 146–155. [Google Scholar] [CrossRef]
  80. Connaughton, V. Glutamate and Glutamate Receptors in the Vertebrate Retina. 1995. Available online: https://webvision.med.utah.edu/book/part-v-phototransduction-in-rods-and-cones/glutamate-and-glutamate-receptors-in-the-vertebrate-retina/ (accessed on 12 January 2023).
  81. Reiner, A.; Levitz, J. Glutamatergic Signaling in the Central Nervous System: Ionotropic and Metabotropic Receptors in Concert. Neuron 2018, 98, 1080–1098. [Google Scholar] [CrossRef]
  82. Hansen, K.B.; Furukawa, H.; Traynelis, S.F. Control of assembly and function of glutamate receptors by the amino-terminal domain. Mol. Pharmacol. 2010, 78, 535–549. [Google Scholar] [CrossRef] [PubMed]
  83. Traynelis, S.F.; Wollmuth, L.P.; McBain, C.J.; Menniti, F.S.; Vance, K.M.; Ogden, K.K.; Hansen, K.B.; Yuan, H.; Myers, S.J.; Dingledine, R. Glutamate Receptor Ion Channels: Structure, Regulation, and Function. Pharmacol. Rev. 2010, 62, 405–496. [Google Scholar] [CrossRef] [PubMed]
  84. Keinänen, K.; Wisden, W.; Sommer, B.; Werner, P.; Herb, A.; Verdoorn, T.A.; Sakmann, B.; Seeburg, P.H. A family of AMPA-selective glutamate receptors. Science 1990, 249, 556–560. [Google Scholar] [CrossRef]
  85. Sommer, B.; Keinänen, K.; Verdoorn, T.A.; Wisden, W.; Burnashev, N.; Herb, A.; Kohler, M.; Takagi, T.; Sakmann, B.; Seeburg, P.H. Flip and flop: A cell-specific functional switch in glutamate-operated channels of the CNS. Science 1990, 249, 1580–1585. [Google Scholar] [CrossRef]
  86. Bleakman, D.; Lodge, D. Neuropharmacology of AMPA and kainate receptors. Neuropharmacology 1998, 37, 1187–1204. [Google Scholar] [CrossRef]
  87. Bardoul, M.; Levallois, C.; König, N. Functional AMPA/kainate receptors in human embryonic and foetal central nervous system. J. Chem. Neuroanat. 1998, 14, 79–85. [Google Scholar] [CrossRef]
  88. Orlandi, C.; Barbon, A.; Barlati, S. Activity regulation of adenosine deaminases acting on RNA (ADARs). Mol. Neurobiol. 2012, 45, 61–75. [Google Scholar] [CrossRef] [PubMed]
  89. Wulff, B.E.; Nishikura, K. Substitutional A-to-I RNA editing. Wiley Interdiscip. Rev. RNA 2010, 1, 90–101. [Google Scholar] [CrossRef] [PubMed]
  90. Kamalova, A.; Nakagawa, T. AMPA receptor structure and auxiliary subunits. J. Physiol. 2021, 599, 453–469. [Google Scholar] [CrossRef]
  91. Noda, M.; Nakanishi, H.; Nabekura, J.; Akaike, N. AMPA-kainate subtypes of glutamate receptor in rat cerebral microglia. J. Neurosci. 2000, 20, 251–258. [Google Scholar] [CrossRef] [PubMed]
  92. Shepherd, J.D.; Huganir, R.L. The cell biology of synaptic plasticity: AMPA receptor trafficking. Annu. Rev. Cell Dev. Biol. 2007, 23, 613–643. [Google Scholar] [CrossRef] [PubMed]
  93. Swanson, G.T.; Kamboj, S.K.; Cull-Candy, S.G. Single-Channel Properties of Recombinant AMPA Receptors Depend on RNA Editing, Splice Variation, and Subunit Composition. J. Neurosci. 1997, 17, 58–69. [Google Scholar] [CrossRef] [PubMed]
  94. Hollmann, M.; Hartley, M.; Heinemann, S. Ca2+ permeability of KA-AMPA—Gated glutamate receptor channels depends on subunit composition. Science 1991, 252, 851–853. [Google Scholar] [CrossRef]
  95. Geiger, J.R.P.; Melcher, T.; Koh, D.-S.; Sakmann, B.; Seeburg, P.; Jonas, P.; Monyer, H. Relative abundance of subunit mRNAs determines gating and Ca2+ permeability of AMPA receptors in principal neurons and interneurons in rat CNS. Neuron 1995, 15, 193–204. [Google Scholar] [CrossRef]
  96. Tomiyama, M.; Rodríguez-Puertas, R.; Cortés, R.; Pazos, A.; Palacios, J.M.; Mengod, G. Flip and flop splice variants of AMPA receptor subunits in the spinal cord of amyotrophic lateral sclerosis. Synapse 2002, 45, 245–249. [Google Scholar] [CrossRef]
  97. Borges, K.; Kettenmann, H. Blockade of K+ channels induced by AMPA/kainate receptor activation in mouse oligodendrocyte precursor cells is mediated by NA+ entry. J. Neurosci. Res. 1995, 42, 579–593. [Google Scholar] [CrossRef] [PubMed]
  98. Ishiuchi, S.; Tsuzuki, K.; Yoshida, Y.; Yamada, N.; Hagimura, N.; Okado, H.; Miwa, A.; Kurihara, H.; Nakazato, Y.; Tamura, M.; et al. Blockage of Ca2+-permeable AMPA receptors suppresses migration and induces apoptosis in human glioblastoma cells. Nat. Med. 2002, 8, 971–978. [Google Scholar] [CrossRef]
  99. Iino, M.; Goto, K.; Kakegawa, W.; Okado, H.; Sudo, M.; Ishiuchi, S.; Miwa, A.; Takayasu, Y.; Saito, I.; Tsuzuki, K.; et al. Glia-synapse interaction through Ca2+-permeable AMPA receptors in Bergmann glia. Science 2001, 292, 926–929. [Google Scholar] [CrossRef]
  100. Fannon, J.; Tarmier, W.; Fulton, D. Neuronal activity and AMPA-type glutamate receptor activation regulates the morphological development of oligodendrocyte precursor cells. Glia 2015, 63, 1021–1035. [Google Scholar] [CrossRef] [PubMed]
  101. Pircher, B.; Pircher, T.; Feigenspan, A. Ionotropic glutamate receptors in the retina—A bioinformatical meta-analysis. bioRxiv 2021. [Google Scholar] [CrossRef]
  102. Bahn, S.; Volk, B.; Wisden, W. Kainate receptor gene expression in the developing rat brain. J. Neurosci. 1994, 14, 5525–5547. [Google Scholar] [CrossRef]
  103. Chittajallu, R.; Vignes, M.; Dev, K.K.; Barnes, J.M.; Collingridge, G.L.; Henley, J.M. Regulation of glutamate release by presynaptic kainate receptors in the hippocampus. Nature 1996, 379, 78–81. [Google Scholar] [CrossRef] [PubMed]
  104. Rodrigues, R.J.; Lerma, J. Metabotropic signaling by kainate receptors. Wiley Interdiscip. Rev. Membr. Transp. Signal. 2012, 1, 399–410. [Google Scholar] [CrossRef]
  105. Rojas, A.; Wetherington, J.; Shaw, R.; Serrano, G.; Swanger, S.; Dingledine, R. Activation of group I metabotropic glutamate receptors potentiates heteromeric kainate receptors. Mol. Pharmacol. 2013, 83, 106–121. [Google Scholar] [CrossRef]
  106. Rojas, A.; Gueorguieva, P.; Lelutiu, N.; Quan, Y.; Shaw, R.; Dingledine, R. The prostaglandin EP1 receptor potentiates kainate receptor activation via a protein kinase C pathway and exacerbates status epilepticus. Neurobiol. Dis. 2014, 70, 74–89. [Google Scholar] [CrossRef] [PubMed]
  107. Rodríguez-Moreno, A.; Sihra, T.S. Metabotropic actions of kainate receptors in the CNS. J. Neurochem. 2007, 103, 2121–2135. [Google Scholar] [CrossRef]
  108. Cull-Candy, S.; Brickley, S.; Farrant, M. NMDA receptor subunits: Diversity, development and disease. Curr. Opin. Neurobiol. 2001, 11, 327–335. [Google Scholar] [CrossRef] [PubMed]
  109. Cull-Candy, S.G.; Leszkiewicz, D.N. Role of distinct NMDA receptor subtypes at central synapses. Sci. STKE 2004, 2004, re16. [Google Scholar] [CrossRef] [PubMed]
  110. Furukawa, H.; Singh, S.K.; Mancusso, R.; Gouaux, E. Subunit arrangement and function in NMDA receptors. Nature 2005, 438, 185–192. [Google Scholar] [CrossRef] [PubMed]
  111. Monyer, H.; Burnashev, N.; Laurie, D.J.; Sakmann, B.; Seeburg, P.H. Developmental and regional expression in the rat brain and functional properties of four NMDA receptors. Neuron 1994, 12, 529–540. [Google Scholar] [CrossRef] [PubMed]
  112. Ciabarra, A.M.; Sullivan, J.M.; Gahn, L.G.; Pecht, G.; Heinemann, S.; Sevarino, K.A. Cloning and characterization of chi-1: A developmentally regulated member of a novel class of the ionotropic glutamate receptor family. J. Neurosci. 1995, 15, 6498–6508. [Google Scholar] [CrossRef]
  113. Sucher, N.J.; Akbarian, S.; Chi, C.; Leclerc, C.; Awobuluyi, M.; Deitcher, D.; Wu, M.; Yuan, J.; Jones, E.; Lipton, S. Developmental and regional expression pattern of a novel NMDA receptor-like subunit (NMDAR-L) in the rodent brain. J. Neurosci. 1995, 15, 6509–6520. [Google Scholar] [CrossRef]
  114. Chatterton, J.E.; Awobuluyi, M.; Premkumar, L.S.; Takahashi, H.; Talantova, M.; Shin, Y.; Cui, J.; Tu, S.; Sevarino, K.A.; Nakanishi, N.; et al. Excitatory glycine receptors containing the NR3 family of NMDA receptor subunits. Nature 2002, 415, 793–798. [Google Scholar] [CrossRef] [PubMed]
  115. Erreger, K.; Chen, P.E.; Wyllie, D.J.A.; Traynelis, S.F. Glutamate receptor gating. Crit. Rev. Neurobiol. 2004, 16, 187–224. [Google Scholar] [CrossRef] [PubMed]
  116. Furukawa, H.; Gouaux, E. Mechanisms of activation, inhibition and specificity: Crystal structures of the NMDA receptor NR1 ligand-binding core. EMBO J. 2003, 22, 2873–2885. [Google Scholar] [CrossRef] [PubMed]
  117. Kim, M.J.; Dunah, A.W.; Wang, Y.T.; Sheng, M. Differential roles of NR2A- and NR2B-containing NMDA receptors in Ras-ERK signaling and AMPA receptor trafficking. Neuron 2005, 46, 745–760. [Google Scholar] [CrossRef]
  118. Strack, S.; Colbran, R.J. Autophosphorylation-dependent targeting of calcium/calmodulin-dependent protein kinase II by the NR2B subunit of the N-methyl-D-aspartate receptor. J. Biol. Chem. 1998, 273, 20689–20692. [Google Scholar] [CrossRef] [PubMed]
  119. Barria, A.; Malinow, R. NMDA receptor subunit composition controls synaptic plasticity by regulating binding to CaMKII. Neuron 2005, 48, 289–301. [Google Scholar] [CrossRef]
  120. Lee, H.K. Synaptic plasticity and phosphorylation. Pharmacol. Ther. 2006, 112, 810–832. [Google Scholar] [CrossRef] [PubMed]
  121. Olloquequi, J.; Cornejo-Córdova, E.; Verdaguer, E.; Soriano, F.X.; Binvignat, O.; Auladell, C.; Camins, A. Excitotoxicity in the pathogenesis of neurological and psychiatric disorders: Therapeutic implications. J. Psychopharmacol. 2018, 32, 265–275. [Google Scholar] [CrossRef] [PubMed]
  122. Crupi, R.; Impellizzeri, D.; Cuzzocrea, S. Role of Metabotropic Glutamate Receptors in Neurological Disorders. Front. Mol. Neurosci. 2019, 12, 20. [Google Scholar] [CrossRef] [PubMed]
  123. Ribeiro, F.M.; Paquet, M.; Cregan, S.P.; Ferguson, S.G. Group I Metabotropic Glutamate Receptor Signalling and its Implication in Neurological Disease. CNS Neurol. Disord. Drug Targets 2010, 9, 574–595. [Google Scholar] [CrossRef] [PubMed]
  124. Lamas, M.; Lee-Rivera, I.; Lòpez-Colomè, A.M. Cell-specific expression of N-methyl-D-aspartate receptor subunits in Müller glia and neurons from the chick retina. Investig. Ophthalmol. Vis. Sci. 2005, 46, 3570–3577. [Google Scholar] [CrossRef] [PubMed]
  125. López, T.; López-Colomé, A.M.; Ortega, A. NMDA receptors in cultured radial glia. FEBS Lett. 1997, 405, 245–248. [Google Scholar] [CrossRef]
  126. López-Colomé, A.M.; López, E.; Mendez-Flores, O.G.; Ortega, A. Glutamate Receptor Stimulation Up-Regulates Glutamate Uptake in Human Müller Glia Cells. Neurochem. Res. 2016, 41, 1797–1805. [Google Scholar] [CrossRef] [PubMed]
  127. Lalo, U.; Pankratov, Y.; Parpura, V.; Verkhratsky, A. Ionotropic receptors in neuronal-astroglial signalling: What is the role of ‘excitable’ molecules in non-excitable cells. Biochim. Biophys. Acta Mol. Cell Res. 2011, 1813, 992–1002. [Google Scholar] [CrossRef]
  128. Clarke, R.J.; Johnson, J.W. Voltage-dependent gating of NR1/2B NMDA receptors. J. Physiol. 2008, 586 Pt 23, 5727–5741. [Google Scholar] [CrossRef] [PubMed]
  129. Mayer, M.L.; Westbrook, G.L.; Guthrie, P.B. Voltage-dependent block by Mg2+ of NMDA responses in spinal cord neurones. Nature 1984, 309, 261–263. [Google Scholar] [CrossRef] [PubMed]
  130. Lalo, U.; Pankratov, Y.; Kirchhoff, F.; North, R.A.; Verkhratsky, A. NMDA receptors mediate neuron-to-glia signaling in mouse cortical astrocytes. J. Neurosci. 2006, 26, 2673–2683. [Google Scholar] [CrossRef]
  131. Verkhratsky, A.; Chvátal, A. NMDA Receptors in Astrocytes. Neurochem. Res. 2020, 45, 122–133. [Google Scholar] [CrossRef] [PubMed]
  132. Verkhratsky, A.; Semyanov, A.; Zorec, R. Physiology of Astroglial Excitability. Function 2020, 1, zqaa016. [Google Scholar] [CrossRef] [PubMed]
  133. Ferraguti, F.; Shigemoto, R. Metabotropic glutamate receptors. Cell Tissue Res. 2006, 326, 483–504. [Google Scholar] [CrossRef] [PubMed]
  134. Sheffler, D.J.; Gregory, K.J.; Rook, J.M.; Conn, P.J. Allosteric Modulation of Metabotropic Glutamate Receptors. Adv. Pharmacol. 2011, 62, 37–77. [Google Scholar] [CrossRef]
  135. Niswender, C.M.; Conn, P.J. Metabotropic glutamate receptors: Physiology, pharmacology, and disease. Annu. Rev. Pharmacol. Toxicol. 2010, 50, 295–322. [Google Scholar] [CrossRef] [PubMed]
  136. Conn, P.J.; Pin, J.P. Pharmacology and functions of metabotropic glutamate receptors. Annu. Rev. Pharmacol. Toxicol. 1997, 37, 205–237. [Google Scholar] [CrossRef] [PubMed]
  137. Rabeh, N.; Hajjar, B.; Maraka, J.O.; Sammanasunathan, A.F.; Khan, M.; Alkhaaldi, S.M.; Mansour, S.; Almheiri, R.T.; Hamdan, H.; Abd-Elrahman, K.S. Targeting mGluR group III for the treatment of neurodegenerative diseases. Biomed. Pharmacother. 2023, 168, 115733. [Google Scholar] [CrossRef]
  138. Suh, Y.H.; Chang, K.; Roche, K.W. Metabotropic glutamate receptor trafficking. Mol. Cell. Neurosci. 2018, 91, 10–24. [Google Scholar] [CrossRef] [PubMed]
  139. Tu, J.C.; Xiao, B.; Naisbitt, S.; Yuan, J.P.; Petralia, R.S.; Brakeman, P.; Doan, A.; Aakalu, V.K.; Lanahan, A.A.; Sheng, M.; et al. Coupling of mGluR/Homer and PSD-95 complexes by the Shank family of postsynaptic density proteins. Neuron 1999, 23, 583–592. [Google Scholar] [CrossRef] [PubMed]
  140. Lan, J.Y.; Skeberdis, V.A.; Jover, T.; Zheng, X.; Bennett, M.V.L.; Zukin, R.S. Activation of Metabotropic Glutamate Receptor 1 Accelerates NMDA Receptor Trafficking. J. Neurosci. 2001, 21, 6058–6068. [Google Scholar] [CrossRef] [PubMed]
  141. Kelly, E.; Bailey, C.P.; Henderson, G. Agonist-selective mechanisms of GPCR desensitization. Br. J. Pharmacol. 2008, 153, S379–S388. [Google Scholar] [CrossRef]
  142. Hanyaloglu, A.C.; Von Zastrow, M. Regulation of GPCRs by endocytic membrane trafficking and its potential implications. Annu. Rev. Pharmacol. Toxicol. 2008, 48, 537–568. [Google Scholar] [CrossRef]
  143. Nelson, C.P.; Challiss, R.A.J. ‘Phenotypic’ pharmacology: The influence of cellular environment on G protein-coupled receptor antagonist and inverse agonist pharmacology. Biochem. Pharmacol. 2007, 73, 737–751. [Google Scholar] [CrossRef]
  144. Ramírez, J.L.; Watt, H.L.; Rocheville, M.; Kumar, U. Agonist-induced up-regulation of human somatostatin receptor type 1 is regulated by β-arrestin-1 and requires an essential serine residue in the receptor C-tail. Biochim. Biophys. Acta 2005, 1669, 182–192. [Google Scholar] [CrossRef] [PubMed]
  145. Achour, L.; Labbé-Jullié, C.; Scott, M.G.H.; Marullo, S. An escort for GPCRs: Implications for regulation of receptor density at the cell surface. Trends Pharmacol. Sci. 2008, 29, 528–535. [Google Scholar] [CrossRef] [PubMed]
  146. Dhami, G.K.; Ferguson, S.S.G. Regulation of metabotropic glutamate receptor signaling, desensitization and endocytosis. Pharmacol. Ther. 2006, 111, 260–271. [Google Scholar] [CrossRef] [PubMed]
  147. Abdul-Ghani, M.A.; Valiante, T.A.; Carlen, P.L.; Pennefather, P.S.; Young, S.R.; Bianchi, R.; Wong, R.K.S.; Sekizawa, S.-I.; Bonham, A.C.; Ireland, D.R.; et al. Metabotropic glutamate receptors coupled to IP3 production mediate inhibition of IAHP in rat dentate granule neurons. J. Neurophysiol. 1996, 76, 2691–2700. [Google Scholar] [CrossRef]
  148. Bhattacharyya, S. Inside story of Group I Metabotropic Glutamate Receptors (mGluRs). Int. J. Biochem. Cell Biol. 2016, 77, 205–212. [Google Scholar] [CrossRef]
  149. Park, H.S.; Betzenhauser, M.J.; Won, J.H.; Chen, J.; Yule, D.I. The type 2 inositol (1,4,5)-trisphosphate (InsP3) receptor determines the sensitivity of InsP3-induced Ca2+ release to ATP in pancreatic acinar cells. J. Biol. Chem. 2008, 283, 26081–26088. [Google Scholar] [CrossRef] [PubMed]
  150. Verkhratsky, A. Physiology and pathophysiology of the calcium store in the endoplasmic reticulum of neurons. Physiol. Rev. 2005, 85, 201–279. [Google Scholar] [CrossRef] [PubMed]
  151. Enz, R. Structure of metabotropic glutamate receptor C-terminal domains in contact with interacting proteins. Front. Mol. Neurosci. 2012, 5, 22883. [Google Scholar] [CrossRef] [PubMed]
  152. Guo, W.; Wei, F.; Zou, S.; Robbins, M.T.; Sugiyo, S.; Ikeda, T.; Tu, J.-C.; Worley, P.F.; Dubner, R.; Ren, K. Group I metabotropic glutamate receptor NMDA receptor coupling and signaling cascade mediate spinal dorsal horn NMDA receptor 2B tyrosine phosphorylation. J. Neurosci. 2004, 24, 9161–9173. [Google Scholar] [CrossRef]
  153. Heidinger, V.; Manzerra, P.; Wang, X.Q.; Strasser, U.; Yu, S.-P.; Choi, D.W.; Behrens, M.M. Metabotropic Glutamate Receptor 1-Induced Upregulation of NMDA Receptor Current: Mediation through the Pyk2/Src-Family Kinase Pathway in Cortical Neurons. J. Neurosci. 2002, 22, 5452–5461. [Google Scholar] [CrossRef]
  154. Minakami, R.; Jinnai, N.; Sugiyama, H. Phosphorylation and calmodulin binding of the metabotropic glutamate receptor subtype 5 (mGluR5) are antagonistic in vitro. J. Biol. Chem. 1997, 272, 20291–20298. [Google Scholar] [CrossRef]
  155. Liu, X.Y.; Chu, X.-P.; Mao, L.-M.; Wang, M.; Lan, H.-X.; Li, M.-H.; Zhang, G.-C.; Parelkar, N.K.; Fibuch, E.E.; Haines, M.; et al. Modulation of D2R-NR2B interactions in response to cocaine. Neuron 2006, 52, 897–909. [Google Scholar] [CrossRef] [PubMed]
  156. Ciruela, F.; Giacometti, A.; McIlhinney, R.A.J. Functional regulation of metabotropic glutamate receptor type 1c: A role for phosphorylation in the desensitization of the receptor. FEBS Lett. 1999, 462, 278–282. [Google Scholar] [CrossRef] [PubMed]
  157. Francesconi, A.; Duvoisin, R.M. Opposing effects of protein kinase C and protein kinase a on metabotropic glutamate receptor signaling: Selective desensitization of the inositol trisphosphate/Ca2+ pathway by phosphorylation of the receptor-G protein-coupling domain. Proc. Natl. Acad. Sci. USA 2000, 97, 6185–6190. [Google Scholar] [CrossRef]
  158. Martínez-Galán, J.R.; López-Bendito, G.; Luján, R.; Shigemoto, R.; Fairén, A.; Valdeolmillos, M. Cajal-Retzius cells in early postnatal mouse cortex selectively express functional metabotropic glutamate receptors. Eur. J. Neurosci. 2001, 13, 1147–1154. [Google Scholar] [CrossRef] [PubMed]
  159. Muñoz, A.; Liu, X.-B.; Jones, E.G. Development of Metabotropic Glutamate Receptors from Trigeminal Nuclei to Barrel Cortex in Postnatal Mouse. J. Comp. Neurol. 1999, 409, 549–566. [Google Scholar] [CrossRef]
  160. Furuta, A.; Martin, L.J. Laminar Segregation of the Cortical Plate During Corticogenesis Is Accompanied by Changes in Glutamate Receptor Expression. J. Neurobiol. 1999, 39, 67–80. [Google Scholar] [CrossRef]
  161. Romano, C.; Van Den Pol, A.N.; O’malley, K.L. Enhanced Early Developmental Expression of the Metabotropic Glutamate Receptor mGluR5 in Rat Brain: Protein, mRNA Splice Variants, and Regional Distribution. J. Comp. Neurol. 1996, 367, 403–412. [Google Scholar] [CrossRef]
  162. Luján, R.; Nusser, Z.; Roberts, J.D.B.; Shigemoto, R.; Somogyi, P. Perisynaptic location of metabotropic glutamate receptors mGluR1 and mGluR5 on dendrites and dendritic spines in the rat hippocampus. Eur. J. Neurosci. 1996, 8, 1488–1500. [Google Scholar] [CrossRef]
  163. Bhave, G.; Karim, F.; Carlton, S.M.; Gereau, R.W., IV. Peripheral group I metabotropic glutamate receptors modulate nociception in mice. Nat. Neurosci. 2001, 4, 417–423. [Google Scholar] [CrossRef]
  164. Jensen, R.J. Activation of group II metabotropic glutamate receptors reduces directional selectivity in retinal ganglion cells. Brain Res. 2006, 1122, 86–92. [Google Scholar] [CrossRef]
  165. Gregory, K.J.; Goudet, C. International union of basic and clinical pharmacology. Cxi. pharmacology, signaling, and physiology of metabotropic glutamate receptors. Pharmacol. Rev. 2021, 73, 521–569. [Google Scholar] [CrossRef]
  166. Mao, L.-M.; Bodepudi, A.; Chu, X.-P.; Wang, J.Q. Group I Metabotropic Glutamate Receptors and Interacting Partners: An Update. Int. J. Mol. Sci. 2022, 23, 840. [Google Scholar] [CrossRef]
  167. Quraishi, S.; Gayet, J.; Morgans, C.W.; Duvoisin, R.M. Distribution of group-III metabotropic glutamate receptors in the retina. J. Comp. Neurol. 2007, 501, 931–943. [Google Scholar] [CrossRef]
  168. Hartveit, E.; Brandstätter, J.H.; Enz, R.; Wässle, H. Expression of the mRNA of seven metabotropic glutamate receptors (mGluR1 to 7) in the rat retina. An in situ hybridization study on tissue sections and isolated cells. Eur. J. Neurosci. 1995, 7, 1472–1483. [Google Scholar] [CrossRef] [PubMed]
  169. Nomura, A.; Shigemoto, R.; Nakamura, Y.; Okamoto, N.; Mizuno, N.; Nakanishi, S. Developmentally regulated postsynaptic localization of a metabotropic glutamate receptor in rat rod bipolar cells. Cell 1994, 77, 361–369. [Google Scholar] [CrossRef]
  170. Brandstätter, J.H.; Koulen, P.; Wässle, H. Diversity of glutamate receptors in the mammalian retina. Vis. Res. 1998, 38, 1385–1397. [Google Scholar] [CrossRef] [PubMed]
  171. Xu, Y.; Dhingra, A.; Fina, M.E.; Koike, C.; Furukawa, T.; Vardi, N. mGluR6 deletion renders the TRPM1 channel in retina inactive. J. Neurophysiol. 2012, 107, 948–957. [Google Scholar] [CrossRef] [PubMed]
  172. Koike, C.; Obara, T.; Uriu, Y.; Numata, T.; Sanuki, R.; Miyata, K.; Koyasu, T.; Ueno, S.; Funabiki, K.; Tani, A.; et al. TRPM1 is a component of the retinal ON bipolar cell transduction channel in the mGluR6 cascade. Proc. Natl. Acad. Sci. USA 2010, 107, 332–337. [Google Scholar] [CrossRef] [PubMed]
  173. Shen, Y.; Rampino, M.A.F.; Carroll, R.C.; Nawy, S. G-protein-mediated inhibition of the Trp channel TRPM1 requires the Gβγ dimer. Proc. Natl. Acad. Sci. USA 2012, 109, 8752–8757. [Google Scholar] [CrossRef] [PubMed]
  174. Rao, A.; Dallman, R.; Henderson, S.; Chen, C.-K. Gβ5 is required for normal light responses and morphology of retinal ON-bipolar cells. J. Neurosci. 2007, 27, 14199–14204. [Google Scholar] [CrossRef] [PubMed]
  175. Zhang, J.; Jeffrey, B.G.; Morgans, C.W.; Burke, N.S.; Haley, T.L.; Duvoisin, R.M.; Brown, R.L. RGS7 and -11 complexes accelerate the ON-bipolar cell light response. Investig. Ophthalmol. Vis. Sci. 2010, 51, 1121–1129. [Google Scholar] [CrossRef] [PubMed]
  176. Vardi, N.; Duvoisin, R.; Wu, G.; Sterling, P. Localization of mGluR6 to Dendrites of ON Bipolar Cells in Primate Retina. J. Comp. Neurol. 2000, 423, 402–412. [Google Scholar] [CrossRef]
  177. Dhingra, A.; Vardi, N. mGlu receptors in the retina. Wiley Interdiscip. Rev. Membr. Transp. Signal. 2012, 1, 641–653. [Google Scholar] [CrossRef]
  178. Masu, M.; Iwakabe, H.; Tagawa, Y.; Miyoshi, T.; Yamashita, M.; Fukuda, Y.; Sasaki, H.; Hiroi, K.; Nakamura, Y.; Shigemoto, R.; et al. Specific deficit of the ON response in visual transmission by targeted disruption of the mGluR6 gene. Cell 1995, 80, 757–765. [Google Scholar] [CrossRef] [PubMed]
  179. Gu, Z.; Liu, W.; Wei, J.; Yan, Z. Regulation of N-methyl-D-aspartic acid (NMDA) receptors by metabotropic glutamate receptor 7. J. Biol. Chem. 2012, 287, 10265–10275. [Google Scholar] [CrossRef]
  180. Rosenberg, N.; Gerber, U.; Ster, J. Activation of Group II Metabotropic Glutamate Receptors Promotes LTP Induction at Schaffer Collateral-CA1 Pyramidal Cell Synapses by Priming NMDA Receptors. J. Neurosci. 2016, 36, 11521–11531. [Google Scholar] [CrossRef] [PubMed]
  181. Joffe, M.E.; Santiago, C.I.; Engers, J.L.; Lindsley, C.W.; Conn, P.J. Metabotropic glutamate receptor subtype 3 gates acute stress-induced dysregulation of amygdalo-cortical function. Mol. Psychiatry 2019, 24, 916–927. [Google Scholar] [CrossRef] [PubMed]
  182. Fei, Z.; Zhang, X.; Bai, H.M.; Jiang, X.F.; Wang, X.L. Metabotropic glutamate receptor antagonists and agonists: Potential neuroprotectors in diffuse brain injury. J. Clin. Neurosci. 2006, 13, 1023–1027. [Google Scholar] [CrossRef]
  183. Folbergrová, J.; Druga, R.; Otáhal, J.; Haugvicová, R.; Mareš, P.; Kubová, H. Seizures induced in immature rats by homocysteic acid and the associated brain damage are prevented by group II metabotropic glutamate receptor agonist (2R,4R)-4-aminopyrrolidine-2,4-dicarboxylate. Exp. Neurol. 2005, 192, 420–436. [Google Scholar] [CrossRef] [PubMed]
  184. Yao, H.; Gao, J.; Feng, Y.B.; Pang, Z.Y.; Chi, Z.F. 2R, 4R-APDC decreases cell proliferation in the dentate gyrus of adult rats: The effect of 2R, 4R-APDC on cell proliferation. Neuroreport 2007, 18, 1459–1462. [Google Scholar] [CrossRef] [PubMed]
  185. Feng, Y.B.; Lin, Y.-T.; Han, Y.-X.; Pang, Y.-J.; Xu, J.-J.; Xue, Y.; Yao, H. 2R,4R-APDC, a Metabotropic Glutamate Receptor Agonist, Reduced Neuronal Apoptosis by Upregulating MicroRNA-128 in a Rat Model After Seizures. Neurochem. Res. 2018, 43, 591–599. [Google Scholar] [CrossRef] [PubMed]
  186. Müller, T.; Möller, T.; Berger, T.; Schnitzer, J.; Kettenmann, H. Calcium Entry Through Kainate Receptors and Resulting Potassium-Channel Blockade in Bergmann Glial Cells. Science 1992, 256, 1563–1566. [Google Scholar] [CrossRef] [PubMed]
  187. Gallo, V.; Ghiani, C.A. Glutamate receptors in glia: New cells, new inputs and new functions. Trends Pharmacol. Sci. 2000, 21, 252–258. [Google Scholar] [CrossRef]
  188. Bouvier, M.; Szatkowski, M.; Amato, A.; Attwell, D. The glial cell glutamate uptake carrier countertransports pH-changing anions. Nature 1992, 360, 471–474. [Google Scholar] [CrossRef] [PubMed]
  189. Puro, D.G. Calcium Channels of Human Retinal Glial Cells. In Methods in Neurosciences; Elsevier: Amsterdam, The Netherlands, 1994; Volume 19, pp. 68–81. [Google Scholar] [CrossRef]
  190. Luque, J.M.; Richards, J.G. Expression of NMDA 2B receptor subunit mRNA in bergmann glia. Glia 1995, 13, 228–232. [Google Scholar] [CrossRef]
  191. Verkhratsky, A.; Kirchhoff, F. NMDA receptors in glia. Neuroscientist 2007, 13, 28–37. [Google Scholar] [CrossRef] [PubMed]
  192. Puro, D.G.; Yuan, J.P.; Sucher, N.J. Activation of NMDA receptor-channels in human retinal Muller glial cells inhibits inward-rectifying potassium currents. Vis. Neurosci. 1996, 13, 319–326. [Google Scholar] [CrossRef]
  193. Mao, L.M.; Geosling, R.; Penman, B.; Wang, J.Q. Local substrates of non-receptor tyrosine kinases at synaptic sites in neurons. Sheng Li Xue Bao 2017, 69, 657. [Google Scholar] [PubMed Central]
  194. Ohnishi, H.; Murata, Y.; Okazawa, H.; Matozaki, T. Src family kinases: Modulators of neurotransmitter receptor function and behavior. Trends Neurosci. 2011, 34, 629–637. [Google Scholar] [CrossRef] [PubMed]
  195. Rajani, V.; Sengar, A.S.; Salter, M.W. Src and Fyn regulation of NMDA receptors in health and disease. Neuropharmacology 2021, 193, 108615. [Google Scholar] [CrossRef] [PubMed]
  196. Bi, X.; Chen, J.; Baudry, M. Calpain-mediated proteolysis of GluR1 subunits in organotypic hippocampal cultures following kainic acid treatment. Brain Res. 1998, 781, 355–357. [Google Scholar] [CrossRef] [PubMed]
  197. Bi, X.; Chang, V.; Molnar, E.; McIlhinney, R.A.J.; Baudry, M. The C-terminal domain of glutamate receptor subunit 1 is a target for calpain-mediated proteolysis. Neuroscience 1996, 73, 903–906. [Google Scholar] [CrossRef] [PubMed]
  198. Bi, X.; Rong, Y.; Chen, J.; Dang, S.; Wang, Z.; Baudry, M. Calpain-mediated regulation of NMDA receptor structure and function. Brain Res. 1998, 790, 245–253. [Google Scholar] [CrossRef] [PubMed]
  199. Vyklický, L. Calcium-mediated modulation of N-methyl-D-aspartate (NMDA) responses in cultured rat hippocampal neurones. J. Physiol. 1993, 470, 575–600. [Google Scholar] [CrossRef] [PubMed]
  200. Ehlers, M.D.; Zhang, S.; Bernhardt, J.P.; Huganir, R.L. Inactivation of NMDA receptors by direct interaction of calmodulin with the NR1 subunit. Cell 1996, 84, 745–755. [Google Scholar] [CrossRef]
  201. Marc, R.E. Kainate activation of horizontal, bipolar, amacrine, and ganglion cells in the rabbit retina. J. Comp. Neurol. 1999, 407, 65–76. [Google Scholar] [CrossRef]
  202. Massey, S.C.; Miller, R.F. Excitatory amino acid receptors of rod- and cone-driven horizontal cells in the rabbit retina. J. Neurophysiol. 1987, 57, 645–659. [Google Scholar] [CrossRef] [PubMed]
  203. Brandstätter, J.H.; Koulen, P.; Kuhn, R.; Van Der Putten, H.; Wässle, H. Compartmental localization of a metabotropic glutamate receptor (mGluR7): Two different active sites at a retinal synapse. J. Neurosci. 1996, 16, 4749–4756. [Google Scholar] [CrossRef]
  204. Fletcher, E.L.; Hack, I.; Brandstätter, J.H.; Wa, H.; Wa, W. Synaptic Localization of NMDA Receptor Subunits in the Rat Retina. J. Comp. Neurol. 2000, 420, 98–112. [Google Scholar] [CrossRef]
  205. Hoffpauir, B.K.; Gleason, E.L. Modulation of synaptic function in retinal amacrine cells. Integr. Comp. Biol. 2005, 45, 658–664. [Google Scholar] [CrossRef]
  206. Peng, Y.W.; Blackstone, C.D.; Huganir, R.L.; Yau, K.W. Distribution of glutamate receptor subtypes in the vertebrate retina. Neuroscience 1995, 66, 483–497. [Google Scholar] [CrossRef]
  207. Cervantes-Villagrana, A.R.; Garcia-Román, J.; Gonzlez-Espinosa, C.; Lamas, M. Pharmacological Inhibition of N-Methyl D-Aspartate Receptor Promotes Secretion of Vascular Endothelial Growth Factor in Müller Cells: Effects of Hyperglycemia and Hypoxia. Curr. Eye Res. 2010, 35, 733–741. [Google Scholar] [CrossRef] [PubMed]
  208. Uchihori, Y.; Puro, D.G. Glutamate as a neuron-to-glial signal for mitogenesis: Role of glial N-methyl-D-aspartate receptors. Brain Res. 1993, 613, 212–220. [Google Scholar] [CrossRef] [PubMed]
  209. Verkhratsky, A.; Trebak, M.; Perocchi, F.; Khananshvili, D.; Sekler, I. Crosslink between calcium and sodium signalling. Exp. Physiol. 2018, 103, 157–169. [Google Scholar] [CrossRef] [PubMed]
  210. Luyt, K.; Váradi, A.; Durant, C.F.; Molnár, E. Oligodendroglial metabotropic glutamate receptors are developmentally regulated and involved in the prevention of apoptosis. J. Neurochem. 2006, 99, 641–656. [Google Scholar] [CrossRef]
  211. Gerber, U.; Gee, C.E.; Benquet, P. Metabotropic glutamate receptors: Intracellular signaling pathways. Curr. Opin. Pharmacol. 2007, 7, 56–61. [Google Scholar] [CrossRef] [PubMed]
  212. Sánchez, G.; Ortega, A. AMPA/KA receptor induced AP-1 DNA binding activity in cultured bergmann glia cells. Neuroreport 1994, 5, 2109–2112. [Google Scholar] [CrossRef] [PubMed]
  213. Puri, B.K. Calcium Signaling and Gene Expression. In Calcium Signaling; Advances in Experimental Medicine and Biology; Springer: Cham, Switzerland, 2020; Volume 1131, pp. 537–545. [Google Scholar] [CrossRef]
  214. Rose, C.R.; Ziemens, D.; Verkhratsky, A. On the special role of NCX in astrocytes: Translating Na+-transients into intracellular Ca2+ signals. Cell Calcium 2020, 86, 102154. [Google Scholar] [CrossRef] [PubMed]
  215. López-Bayghen, E.; Ortega, A. Glutamate-dependent transcriptional regulation of GLAST: Role of PKC. J. Neurochem. 2004, 91, 200–209. [Google Scholar] [CrossRef] [PubMed]
  216. Barrera, I.; Flores-Méndez, M.; Hernández-Kelly, L.C.; Cid, L.; Huerta, M.; Zinker, S.; López-Bayghen, E.; Aguilera, J.; Ortega, A. Glutamate regulates eEF1A phosphorylation and ribosomal transit time in Bergmann glial cells. Neurochem. Int. 2010, 57, 795–803. [Google Scholar] [CrossRef] [PubMed]
  217. Martínez-Lozada, Z.; Hernández-Kelly, L.C.; Aguilera, J.; López-Bayghen, E.; Ortega, A. Signaling through EAAT-1/GLAST in cultured Bergmann glia cells. Neurochem. Int. 2011, 59, 871–879. [Google Scholar] [CrossRef] [PubMed]
  218. López-Bayghen, E.; Espinoza-Rojo, M.; Ortega, A. Glutamate down-regulates GLAST expression through AMPA receptors in Bergmann glial cells. Mol. Brain Res. 2003, 115, 1–9. [Google Scholar] [CrossRef]
  219. Aguirre, A.; López-Bayghen, E.; Ortega, A. Glutamate-dependent transcriptional regulation of the chkbp gene: Signaling mechanisms. J. Neurosci. Res. 2002, 70, 117–127. [Google Scholar] [CrossRef]
  220. Chai, R.; Xing, C.; Gao, D.; Yuan, H.; Zhan, Y.; Wang, S. Remote-Controlling Potassium Channels in Living Cells through Photothermal Inactivation of Calmodulin. Adv. Healthc. Mater. 2018, 7, 1800674. [Google Scholar] [CrossRef]
  221. Wyttenbach, T.; Grabenauer, M.; Thalassinos, K.; Scrivens, J.H.; Bowers, M.T. The effect of calcium ions and peptide ligands on the relative stabilities of the calmodulin dumbbell and compact structures. J. Phys. Chem. B 2010, 114, 437–447. [Google Scholar] [CrossRef]
  222. Kazanietz, M.G. Novel ‘nonkinase’ phorbol ester receptors: The C1 domain connection. Mol. Pharmacol. 2002, 61, 759–767. [Google Scholar] [CrossRef]
  223. Millán, A.; Arias-Montaño, J.A.; Méndez, J.A.; Hernández-Kelly, L.C.R.; Ortega, A. α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors signaling complexes in Bergmann glia. J. Neurosci. Res. 2004, 78, 56–63. [Google Scholar] [CrossRef]
  224. López-Bayghen, E.; Aguirre, A.; Ortega, A. Transcriptional regulation through glutamate receptors: Involvement of tyrosine kinases. J. Neurosci. Res. 2003, 74, 717–725. [Google Scholar] [CrossRef]
  225. Aguirre, A.; López, T.; López-Bayghen, E.; Ortega, A. Glutamate regulates kainate-binding protein expression in cultured chick Bergmann glia through an activator protein-1 binding site. J. Biol. Chem. 2000, 275, 39246–39253. [Google Scholar] [CrossRef] [PubMed]
  226. Millán, A.; Aguilar, P.; Méndez, J.A.; Arias-Montaño, J.A.; Ortega, A. Glutamate activates PP125FAK through AMPA/kainate receptors in Bergmann glia. J. Neurosci. Res. 2001, 66, 723–729. [Google Scholar] [CrossRef] [PubMed]
  227. Rosas, S.; Vargas, M.A.; López-Bayghen, E.; Ortega, A. Glutamate-dependent transcriptional regulation of GLAST/EAAT1: A role for YY1. J. Neurochem. 2007, 101, 1134–1144. [Google Scholar] [CrossRef] [PubMed]
  228. Poblete-Naredo, I.; Angulo, C.; Hernández-Kelly, L.; López-Bayghen, E.; Aguilera, J.; Ortega, A. Insulin-dependent regulation of GLAST/EAAT1 in Bergmann glial cells. Neurosci. Lett. 2009, 451, 134–138. [Google Scholar] [CrossRef] [PubMed]
  229. López-Colomé, A.M.; Murbartián, J.; Ortega, A. Excitatory amino acid-induced AP-1 DNA binding activity in Müller glia. J. Neurosci. Res. 1995, 41, 179–184. [Google Scholar] [CrossRef] [PubMed]
  230. López-Colomé, A.M.; Ortega, A.; Romo-De-Vivar, M. Excitatory amino acid-induced phosphoinositide hydrolysis in Müller glia. Glia 1993, 9, 127–135. [Google Scholar] [CrossRef] [PubMed]
  231. Stevens, E.R.; Esguerra, M.; Kim, P.M.; Newman, E.A.; Snyder, S.H.; Zahs, K.R.; Miller, R.F. D-serine and serine racemase are present in the vertebrate retina and contribute to the physiological activation of NMDA receptors. Proc. Natl. Acad. Sci. USA 2003, 100, 6789–6794. [Google Scholar] [CrossRef] [PubMed]
  232. Sullivan, S.M.; Lee, A.; Björkman, S.T.; Miller, S.M.; Sullivan, R.K.P.; Poronnik, P.; Colditz, P.B.; Pow, D.V. Cytoskeletal anchoring of GLAST determines susceptibility to brain damage: An identified role for GFAP. J. Biol. Chem. 2007, 282, 29414–29423. [Google Scholar] [CrossRef]
  233. Hellyer, S.; Leach, K.; Gregory, K.J. Neurobiological insights and novel therapeutic opportunities for CNS disorders from mGlu receptor allosteric and biased modulation. Curr. Opin. Pharmacol. 2017, 32, 49–55. [Google Scholar] [CrossRef] [PubMed]
  234. Durand, D.; Carniglia, L.; Turati, J.; Ramírez, D.; Saba, J.; Caruso, C.; Lasaga, M. Amyloid-beta neurotoxicity and clearance are both regulated by glial group II metabotropic glutamate receptors. Neuropharmacology 2017, 123, 274–286. [Google Scholar] [CrossRef] [PubMed]
  235. Ribeiro, F.M.; Vieira, L.B.; Pires, R.G.W.; Olmo, R.P.; Ferguson, S.S.G. Metabotropic glutamate receptors and neurodegenerative diseases. Pharmacol. Res. 2017, 115, 179–191. [Google Scholar] [CrossRef] [PubMed]
  236. Caraci, F.; Molinaro, G.; Battaglia, G.; Giuffrida, M.L.; Riozzi, B.; Traficante, A.; Bruno, V.; Cannella, M.; Merlo, S.; Wang, X.; et al. Targeting Group II Metabotropic Glutamate (mGlu) Receptors for the Treatment of Psychosis Associated with Alzheimer’s Disease: Selective Activation of mGlu2 Receptors Amplifies β-Amyloid Toxicity in Cultured Neurons, Whereas Dual Activation of mGlu2 and mGlu3 Receptors Is Neuroprotective. Mol. Pharmacol. 2011, 79, 618–626. [Google Scholar] [CrossRef]
  237. Zhang, H.; Xue, J.; Hessler, P.; Tahir, S.K.; Chen, J.; Jin, S.; Souers, A.J.; Leverson, J.D.; Lam, L.T. Genomic analysis and selective small molecule inhibition identifies BCL-XL as a critical survival factor in a subset of colorectal cancer. Mol. Cancer 2015, 14, 126. [Google Scholar] [CrossRef] [PubMed]
  238. Jantas, D.; Lech, T.; Gołda, S.; Pilc, A.; Lasoń, W. New evidences for a role of mGluR7 in astrocyte survival: Possible implications for neuroprotection. Neuropharmacology 2018, 141, 223–237. [Google Scholar] [CrossRef] [PubMed]
  239. Aronica, E.; Gorter, J.A.; Ijlst-Keizers, H.; Rozemuller, A.J.; Yankaya, B.; Leenstra, S.; Troost, D. Expression and functional role of mGluR3 and mGluR5 in human astrocytes and glioma cells: Opposite regulation of glutamate transporter proteins. Eur. J. Neurosci. 2003, 17, 2106–2118. [Google Scholar] [CrossRef] [PubMed]
  240. Gegelashvili, G.; Dehnes, Y.; Danbolt, N.C.; Schousboe, A. The high-affinity glutamate transporters GLT1, GLAST, and EAAT4 are regulated via different signalling mechanisms. Neurochem. Int. 2000, 37, 163–170. [Google Scholar] [CrossRef] [PubMed]
  241. Lin, C.H.; You, J.R.; Wei, K.C.; Gean, P.W. Stimulating ERK/PI3K/NFκB signaling pathways upon activation of mGluR2/3 restores OGD-induced impairment in glutamate clearance in astrocytes. Eur. J. Neurosci. 2014, 39, 83–96. [Google Scholar] [CrossRef] [PubMed]
  242. Smith, S.B. Diabetic retinopathy and the NMDA receptor. Drug News Perspect. 2002, 15, 226–232. [Google Scholar] [CrossRef]
  243. Vorwerk, C.K.; Gorla, M.S.R.; Dreyer, E.B. An Experimental Basis for Implicating Excitotoxicity in Glaucomatous Optic Neuropathy. Surv. Ophthalmol. 1999, 43, S142–S150. [Google Scholar] [CrossRef] [PubMed]
  244. Gowda, K.; Zinnantif, W.J.; LaNoue, K.F. The influence of diabetes on glutamate metabolism in retinas. J. Neurochem. 2011, 117, 309–320. [Google Scholar] [CrossRef] [PubMed]
  245. Coyle, J.T.; Puttfarcken, P. Oxidative stress, glutamate, and neurodegenerative disorders. Science 1993, 262, 689–695. [Google Scholar] [CrossRef] [PubMed]
  246. Waggie, K.S.; Kahle, P.J.; Tolwani, R.J. Neurons and mechanisms of neuronal death in neurodegenerative diseases: A brief review. Lab. Anim. Sci. 1999, 49, 358–362. [Google Scholar] [PubMed]
  247. Opere, C.A.; Heruye, S.; Njie-Mbye, Y.F.; Ohia, S.E.; Sharif, N.A. Regulation of excitatory amino acid transmission in the retina: Studies on neuroprotection. J. Ocul. Pharmacol. Ther. 2018, 34, 107–118. [Google Scholar] [CrossRef] [PubMed]
  248. Xia, M.; Zhang, F. Amino Acids Metabolism in Retinopathy: From Clinical and Basic Research Perspective. Metabolites 2022, 12, 1244. [Google Scholar] [CrossRef]
  249. Berliocchi, L.; Bano, D.; Nicotera, P. Ca2+ signals and death programmes in neurons. Philos. Trans. R. Soc. B Biol. Sci. 2005, 360, 2255–2258. [Google Scholar] [CrossRef]
  250. Hartwick, A.T.E.; Hamilton, C.M.; Baldridge, W.H. Glutamatergic calcium dynamics and deregulation of rat retinal ganglion cells. J. Physiol. 2008, 586, 3425–3446. [Google Scholar] [CrossRef]
  251. Santiago, A.R.; Hughes, J.M.; Kamphuis, W.; Schlingemann, R.O.; Ambrósio, A.F. Diabetes changes ionotropic glutamate receptor subunit expression level in the human retina. Brain Res. 2008, 1198, 153–159. [Google Scholar] [CrossRef] [PubMed]
  252. Baudry, M.; Bi, X.; Gall, C.; Lynch, G. The biochemistry of memory: The 26 year journey of a ‘new and specific hypothesis’. Neurobiol. Learn. Mem. 2011, 95, 125–133. [Google Scholar] [CrossRef] [PubMed]
  253. Lee, Y.S.; Silva, A.J. The molecular and cellular biology of enhanced cognition. Nat. Rev. Neurosci. 2009, 10, 126–140. [Google Scholar] [CrossRef] [PubMed]
  254. Friedman, L.K. Calcium: A role for neuroprotection and sustained adaptation. Mol. Interv. 2006, 6, 315–329. [Google Scholar] [CrossRef]
  255. König, N.; Poluch, S.; Estabel, J.; Durand, M.; Drian, M.J.; Exbrayat, J.M. Synaptic and non-synaptic AMPA receptors permeable to calcium. Jpn. J. Pharmacol. 2001, 86, 1–17. [Google Scholar] [CrossRef]
  256. Kelly, L.; Brown, C.; Gibbard, A.G.; Jackson, T.; Swinny, J.D. Subunit-specific expression and function of AMPA receptors in the mouse locus coeruleus. Am. J. Anat. 2023, 243, 813–825. [Google Scholar] [CrossRef]
  257. Gorter, J.A.; Petrozzino, J.J.; Aronica, E.M.; Rosenbaum, D.M.; Opitz, T.; Bennett, M.V.L.; Connor, J.A.; Zukin, R.S. Global ischemia induces downregulation of Glur2 mRNA and increases AMPA receptor-mediated Ca2+ influx in hippocampal CA1 neurons of gerbil. J. Neurosci. 1997, 17, 6179–6188. [Google Scholar] [CrossRef] [PubMed]
  258. Noh, K.M.; Yokota, H.; Mashiko, T.; Castillo, P.E.; Zukin, R.S.; Bennett, M.V.L. Blockade of calcium-permeable AMPA receptors protects hippocampal neurons against global ischemia-induced death. Proc. Natl. Acad. Sci. USA 2005, 102, 12230–12235. [Google Scholar] [CrossRef] [PubMed]
  259. Guo, C.; Ma, Y.Y. Calcium Permeable-AMPA Receptors and Excitotoxicity in Neurological Disorders. Front. Neural Circuits 2021, 15, 711564. [Google Scholar] [CrossRef] [PubMed]
  260. Schrammel, A.; Gorren, A.C.F.; Schmidt, K.; Pfeiffer, S.; Mayer, B. S-nitrosation of glutathione by nitric oxide, peroxynitrite, and NO/O2•−. Free Radic. Biol. Med. 2003, 34, 1078–1088. [Google Scholar] [CrossRef] [PubMed]
  261. Brown, G.C. Nitric oxide and neuronal death. Nitric Oxide 2010, 23, 153–165. [Google Scholar] [CrossRef] [PubMed]
  262. Kritis, A.A.; Stamoula, E.G.; Paniskaki, K.A.; Vavilis, T.D. Researching glutamate—Induced cytotoxicity in different cell lines: A comparative/collective analysis/study. Front. Cell Neurosci. 2015, 9, 91. [Google Scholar] [CrossRef] [PubMed]
  263. Tanaka, S.; Fujio, Y.; Nakayama, H. Caveolae-Specific CaMKII Signaling in the Regulation of Voltage-Dependent Calcium Channel and Cardiac Hypertrophy. Front. Physiol. 2018, 9, 1081. [Google Scholar] [CrossRef] [PubMed]
  264. Brewster, A.L.; Marzec, K.; Hairston, A.; Ho, M.; Anderson, A.E.; Lai, Y.-C. Early cardiac electrographic and molecular remodeling in a model of status epilepticus and acquired epilepsy. Epilepsia 2016, 57, 1907–1915. [Google Scholar] [CrossRef] [PubMed]
  265. Cooper, N.G.F.; Laabich, A.; Fan, W.; Wang, X. The relationship between neurotrophic factors and CaMKII in the death and survival of retinal ganglion cells. Prog. Brain Res. 2008, 173, 521–540. [Google Scholar] [CrossRef] [PubMed]
  266. Zhou, K.; Enkhjargal, B.; Xie, Z.; Sun, C.; Wu, L.; Malaguit, J.; Chen, S.; Tang, J.; Zhang, J.; Zhang, J.H. Dihydrolipoic acid inhibits lysosomal rupture and NLRP3 through lysosome-associated membrane Protein-1/Calcium/Calmodulin-Dependent Protein Kinase II/TAK1 pathways after subarachnoid hemorrhage in rat. Stroke 2018, 49, 175–183. [Google Scholar] [CrossRef] [PubMed]
  267. Qian, Y.; Xia, T.; Cui, Y.; Chu, S.; Ma, Z.; Gu, X. The role of CaMKII in neuropathic pain and fear memory in chronic constriction injury in rats. Int. J. Neurosci. 2019, 129, 146–154. [Google Scholar] [CrossRef]
  268. Wang, S.; Liao, L.; Huang, Y.; Wang, M.; Zhou, H.; Chen, D.; Liu, F.; Ji, D.; Xia, X.; Jiang, B.; et al. Pin1 is regulated by CaMKII activation in glutamate-induced retinal neuronal regulated necrosis. Front. Cell. Neurosci. 2019, 13, 276. [Google Scholar] [CrossRef]
  269. Beyersmann, D.; Haase, H. Functions of zinc in signaling, proliferation and differentiation of mammalian cells. BioMetals 2001, 14, 331–341. [Google Scholar] [CrossRef]
  270. Jia, Y.; Jeng, J.M.; Sensi, S.L.; Weiss, J.H. Zn2+ currents are mediated by calcium-permeable AMPA/kainate channels in cultured murine hippocampal neurones. J. Physiol. 2002, 543 Pt 1, 35–48. [Google Scholar] [CrossRef]
  271. Van Damme, P.; Braeken, D.; Callewaert, G.; Robberecht, W.; Van Den Bosch, L. GluR2 deficiency accelerates motor neuron degeneration in a mouse model of amyotrophic lateral sclerosis. J. Neuropathol. Exp. Neurol. 2005, 64, 605–612. [Google Scholar] [CrossRef] [PubMed]
  272. Van Damme, P.; Bogaert, E.; Dewil, M.; Hersmus, N.; Kiraly, D.; Scheveneels, W.; Bockx, I.; Braeken, D.; Verpoorten, N.; Verhoeven, K.; et al. Astrocytes regulate GluR2 expression in motor neurons and their vulnerability to excitotoxicity. Proc. Natl. Acad. Sci. USA 2007, 104, 14825–14830. [Google Scholar] [CrossRef]
  273. Santos, A.E.; Duarte, C.B.; Iizuka, M.; Barsoumian, E.L.; Ham, J.; Lopes, M.C.; Carvalho, A.L. Excitotoxicity mediated by Ca2+-permeable GluR4-containing AMPA receptors involves the AP-1 transcription factor. Cell Death Differ. 2006, 13, 652–660. [Google Scholar] [CrossRef] [PubMed]
  274. Siliprandi, R.; Lipartiti, M.; Fadda, E.; Arban, R.; Kozikowski, A.P.; Manev, H. A derivative of a rigid glutamate analog protects the retina from excitotoxicity. Neuroreport 1994, 5, 1227–1229. [Google Scholar] [CrossRef]
  275. Hlaváčková, M.; Kožichová, K.; Neckář, J.; Kolář, F.; Musters, R.J.P.; Novák, F.; Nováková, O. Up-regulation and redistribution of protein kinase C-δ in chronically hypoxic heart. Mol. Cell. Biochem. 2010, 345, 271–282. [Google Scholar] [CrossRef] [PubMed]
  276. Casson, R.J.; Franzco, D. Possible role of excitotoxicity in the pathogenesis of glaucoma. Clin. Exp. Ophthalmol. 2006, 34, 54–63. [Google Scholar] [CrossRef]
  277. Wang, X.; Mo, X.; Li, D.; Wang, Y.; Fang, Y.; Rong, X.; Miao, H.; Shou, T. Neuroprotective effect of transcorneal electrical stimulation on ischemic damage in the rat retina. Exp. Eye Res. 2011, 93, 753–760. [Google Scholar] [CrossRef] [PubMed]
  278. Quigley, H.A.; Nickells, R.W.; Kerrigan, L.A.; Pease, M.E.; Thibault, D.J.; Zack, D.J. Retinal ganglion cell death in experimental glaucoma and after axotomy occurs by apoptosis. Investig. Ophthalmol. Vis. Sci. 1995, 36, 774–786. [Google Scholar]
  279. Schor, N.F. Inactivation of mammalian brain glutamine synthetase by oxygen radicals. Brain Res. 1988, 456, 17–21. [Google Scholar] [CrossRef] [PubMed]
  280. Prentice, H.; Modi, J.P.; Wu, J.Y. Mechanisms of Neuronal Protection against Excitotoxicity, Endoplasmic Reticulum Stress, and Mitochondrial Dysfunction in Stroke and Neurodegenerative Diseases. Oxid. Med. Cell. Longev. 2015, 2015, 964518. [Google Scholar] [CrossRef]
  281. Doyle, K.P.; Simon, R.P.; Stenzel-Poore, M.P. Mechanisms of ischemic brain damage. Neuropharmacology 2008, 55, 310–318. [Google Scholar] [CrossRef] [PubMed]
  282. Rossi, D.J.; Oshima, T.; Attwell, D. Glutamate release in severe brain ischaemia is mainly by reversed uptake. Nature 2000, 403, 316–321. [Google Scholar] [CrossRef]
  283. Attwell, D.; Laughlin, S.B. An energy budget for signaling in the grey matter of the brain. J. Cereb. Blood Flow Metab. 2001, 21, 1133–1145. [Google Scholar] [CrossRef]
  284. Erecinska, M.; Silver, I.A. ATP and brain function. J. Cereb. Blood Flow Metab. 1989, 9, 2–19. [Google Scholar] [CrossRef]
  285. Farhat, E.; Devereaux, M.E.M.; Cheng, H.; Weber, J.-M.; Pamenter, M.E. Na+/K+-ATPase activity is regionally regulated by acute hypoxia in naked mole-rat brain. Neurosci. Lett. 2021, 764, 136244. [Google Scholar] [CrossRef]
  286. Kirdajova, D.B.; Kriska, J.; Tureckova, J.; Anderova, M. Ischemia-Triggered Glutamate Excitotoxicity from the Perspective of Glial Cells. Front. Cell. Neurosci. 2020, 14, 51. [Google Scholar] [CrossRef] [PubMed]
  287. Armada-Moreira, A.; Gomes, J.I.; Pina, C.C.; Savchak, O.K.; Gonçalves-Ribeiro, J.; Rei, N.; Pinto, S.; Morais, T.P.; Martins, R.S.; Ribeiro, F.F.; et al. Going the Extra (Synaptic) Mile: Excitotoxicity as the Road Toward Neurodegenerative Diseases. Front. Cell. Neurosci. 2020, 14, 90. [Google Scholar] [CrossRef] [PubMed]
  288. Chua, J.; Nivison-Smith, L.; Fletcher, E.L.; Trenholm, S.; Awatramani, G.B.; Kalloniatis, M. Early remodeling of Müller cells in the rd/rd mouse model of retinal dystrophy. J. Comp. Neurol. 2013, 521, 2439–2453. [Google Scholar] [CrossRef] [PubMed]
  289. Jones, B.W.; Pfeiffer, R.L.; Ferrell, W.D.; Watt, C.B.; Marmor, M.; Marc, R.E. Retinal remodeling in human retinitis pigmentosa. Exp. Eye Res. 2016, 150, 149–165. [Google Scholar] [CrossRef]
  290. Pow, D.V.; Robinson, S.R. Glutamate in some retinal neurons is derived solely from glia. Neuroscience 1994, 60, 355–366. [Google Scholar] [CrossRef] [PubMed]
  291. Bringmann, A.; Grosche, A.; Pannicke, T.; Reichenbach, A. GABA and Glutamate Uptake and Metabolism in Retinal Glial (Müller) Cells. Front. Endocrinol. 2013, 4, 48. [Google Scholar] [CrossRef] [PubMed]
  292. Gorovits, R.; Avidan, N.; Avisar, N.; Shaked, I.; Vardimon, L. Glutamine synthetase protects against neuronal degeneration in injured retinal tissue. Proc. Natl. Acad. Sci. USA 1997, 94, 7024–7029. [Google Scholar] [CrossRef] [PubMed]
  293. Giugliano, D.; Ceriello, A.; Paolisso, G. Diabetes mellitus, hypertension, and cardiovascular disease: Which role for oxidative stress? Metabolism 1995, 44, 363–368. [Google Scholar] [CrossRef] [PubMed]
  294. Yen, M.Y.; Wang, A.G.; Wei, Y.H. Leber’s hereditary optic neuropathy: A multifactorial disease. Prog. Retin. Eye Res. 2006, 25, 381–396. [Google Scholar] [CrossRef] [PubMed]
  295. Newman, E.A. Inward-rectifying potassium channels in retinal glial (müller) cells. J. Neurosci. 1993, 13, 3333–3345. [Google Scholar] [CrossRef] [PubMed]
  296. Vinores, S.A. Breakdown of the Blood–Retinal Barrier. Encycl. Eye 2010, 216–222. [Google Scholar] [CrossRef]
  297. Fleegal, M.A.; Hom, S.; Borg, L.K.; Davis, T.P. Activation of PKC modulates blood-brain barrier endothelial cell permeability changes induced by hypoxia and posthypoxic reoxygenation. Am. J. Physiol. Heart Circ. Physiol. 2005, 289, H2012–H2019. [Google Scholar] [CrossRef] [PubMed]
  298. Sun, M.K.; Alkon, D.L. Activation of protein kinase C isozymes for the treatment of dementias. Adv. Pharmacol. 2012, 64, 273–302. [Google Scholar] [CrossRef]
  299. Aloulou, A.; Rahier, R.; Arhab, Y.; Noiriel, A.; Abousalham, A. Phospholipases: An Overview. In Lipases and Phospholipases; Methods in Molecular Biology; Humana Press: New York, NY, USA, 2018; Volume 1835, pp. 69–105. [Google Scholar] [CrossRef]
  300. Lin, Y.; Jones, B.W.; Liu, A.; Vazquéz-Chona, F.R.; Lauritzen, J.S.; Ferrell, W.D.; Marc, R.E. Rapid glutamate receptor 2 trafficking during retinal degeneration. Mol. Neurodegener. 2012, 7, 7. [Google Scholar] [CrossRef] [PubMed]
  301. Tan, T.E.; Wong, T.Y. Diabetic retinopathy: Looking forward to 2030. Front. Endocrinol. 2023, 13, 1077669. [Google Scholar] [CrossRef] [PubMed]
  302. Rodríguez-Campuzano, A.G.; Hernández-Kelly, L.C.; Ortega, A. DNA Methylation-Dependent Gene Expression Regulation of Glutamate Transporters in Cultured Radial Glial Cells. Mol. Neurobiol. 2022, 59, 1912–1924. [Google Scholar] [CrossRef] [PubMed]
  303. Wu, J.; Liu, L.-L.; Cao, M.; Hu, A.; Hu, D.; Luo, Y.; Wang, H.; Zhong, J.-N. DNA methylation plays important roles in retinal development and diseases. Exp. Eye Res. 2021, 211, 108733. [Google Scholar] [CrossRef] [PubMed]
  304. De Groot, J.F.; Ta, J.L.; Fuller, G.; Yung, W.K.A. The excitatory amino acid transporter-2 induces apoptosis and decreases glioma growth in vitro and in vivo. Cancer Res. 2005, 65, 1934–1940. [Google Scholar] [CrossRef]
  305. Okabe, Y.; Takahashi, T.; Mitsumasu, C.; Kosai, K.I.; Tanaka, E.; Matsuishi, T. Alterations of gene expression and glutamate clearance in astrocytes derived from an mecp2-null mouse model of rett syndrome. PLoS ONE 2012, 7, e35354. [Google Scholar] [CrossRef] [PubMed]
  306. Dunn, H.G.; MacLeod, P.M. Rett syndrome: Review of biological abnormalities. Can. J. Neurol. Sci. 2001, 28, 16–29. [Google Scholar] [CrossRef] [PubMed]
  307. Guy, J.; Hendrich, B.; Holmes, M.; Martin, J.E.; Bird, A. A mouse Mecp2-null mutation causes neurological symptoms that mimic Rett syndrome. Nat. Genet. 2001, 27, 322–326. [Google Scholar] [CrossRef]
  308. Bruno, V.; Battaglia, G.; Copani, A.; Giffard, R.G.; Raciti, G.; Raffaele, R.; Shinozaki, H.; Nicoletti, F. Activation of Class II or III Metabotropic Glutamate Receptors Protects Cultured Cortical Neurons Against Excitotoxic Degeneration. Eur. J. Neurosci. 1995, 7, 1906–1913. [Google Scholar] [CrossRef]
  309. Flor, P.J.; Battaglia, G.; Nicoletti, F.; Gasparini, F.; Bruno, V. Neuroprotective Activity of Metabotropic Glutamate Receptor Ligands. 2013. Available online: https://www.ncbi.nlm.nih.gov/books/NBK6556/ (accessed on 6 January 2024).
  310. Kingston, A.E.; O’Neill, M.J.; Lam, A.; Bales, K.R.; Monn, J.A.; Schoepp, D.D. Neuroprotection by metabotropic glutamate receptor agonists: LY354740, LY379268 and LY389795. Eur. J. Pharmacol. 1999, 377, 155–165. [Google Scholar] [CrossRef]
  311. Spampinato, S.F.; Copani, A.; Nicoletti, F.; Sortino, M.A.; Caraci, F. Metabotropic glutamate receptors in glial cells: A new potential target for neuroprotection? Front. Mol. Neurosci. 2018, 11, 414. [Google Scholar] [CrossRef]
  312. Calabresi, P.; Centonze, D.; Cupini, L.M.; Costa, C.; Pisani, F.; Bernardi, G. Ionotropic glutamate receptors: Still a target for neuroprotection in brain ischemia? Insights from in vitro studies. Neurobiol. Dis. 2003, 12, 82–88. [Google Scholar] [CrossRef]
  313. Li, Y.; Li, F.; Qin, D.; Chen, H.; Wang, J.; Wang, J.; Song, S.; Wang, C.; Wang, Y.; Liu, S.; et al. The role of brain derived neurotrophic factor in central nervous system. Front. Aging Neurosci. 2022, 14, 986443. [Google Scholar] [CrossRef]
Figure 1. Overview of Glu-mediated vertical pathways in the retina. In the visual system, light signals are transmitted through two main pathways: the horizontal pathway and the vertical pathway. The vertical pathway involves photoreceptors, bipolar cells, and retinal ganglion cells, which transmit signals to the brain. Glu is the primary neurotransmitter in this pathway, acting on two types of receptors: GRI (GRIA, GRIN, and GRIK) and GRM (GRM1-8). Glu receptors are abundantly expressed both on the neuronal and MCs. These receptors’ activities are regulated by various factors, including subunit composition, density, affinity, and localization. Glu receptor expression on neurons is involved in important neuronal processes. Müller glial cells spread across the retinal layers, forming close interaction with the retinal neurons, in what is known as a tripartite synapse. After Glu uptake by high-affinity transporters located abundantly on MCs, Glu is converted rapidly into Gln by the Gln synthetase enzyme. Effective coordination between Glu uptake and degradation is essential for maintaining a functional Glu/Gln cycle. Gln is released by MCs into the extra-synaptic space presumably through Sodium dependent transporter 3 (SNAT3) thereafter, Gln is taken up into the presynaptic neurons by SNAT2, where it undergoes transformation into Glu by the activity of phosphate-activated glutaminase (PAG). This metabolic pathway appears to serve as the primary source of Glu in retinal bipolar and ganglion cells. Following synthesis, Glu is shuttled into presynaptic vesicles via a specialized transporter vGLUT. This transport process is facilitated by the membrane potential generated by the vacuolar H+-ATPase (V-ATPase) across the vesicular membrane, ensuring high concentrations of Glu within the vesicles.
Figure 1. Overview of Glu-mediated vertical pathways in the retina. In the visual system, light signals are transmitted through two main pathways: the horizontal pathway and the vertical pathway. The vertical pathway involves photoreceptors, bipolar cells, and retinal ganglion cells, which transmit signals to the brain. Glu is the primary neurotransmitter in this pathway, acting on two types of receptors: GRI (GRIA, GRIN, and GRIK) and GRM (GRM1-8). Glu receptors are abundantly expressed both on the neuronal and MCs. These receptors’ activities are regulated by various factors, including subunit composition, density, affinity, and localization. Glu receptor expression on neurons is involved in important neuronal processes. Müller glial cells spread across the retinal layers, forming close interaction with the retinal neurons, in what is known as a tripartite synapse. After Glu uptake by high-affinity transporters located abundantly on MCs, Glu is converted rapidly into Gln by the Gln synthetase enzyme. Effective coordination between Glu uptake and degradation is essential for maintaining a functional Glu/Gln cycle. Gln is released by MCs into the extra-synaptic space presumably through Sodium dependent transporter 3 (SNAT3) thereafter, Gln is taken up into the presynaptic neurons by SNAT2, where it undergoes transformation into Glu by the activity of phosphate-activated glutaminase (PAG). This metabolic pathway appears to serve as the primary source of Glu in retinal bipolar and ganglion cells. Following synthesis, Glu is shuttled into presynaptic vesicles via a specialized transporter vGLUT. This transport process is facilitated by the membrane potential generated by the vacuolar H+-ATPase (V-ATPase) across the vesicular membrane, ensuring high concentrations of Glu within the vesicles.
Receptors 04 00004 g001
Figure 2. Glu receptors classification. Glu receptors are broadly divided into GRI and GRM. These receptors are distinguished based on structure, binding affinities, and signaling. On the left, the table illustrates GRI subunits and their corresponding coding genes. The proteins and coding genes belonging to the GRM families are depicted on the right panel.
Figure 2. Glu receptors classification. Glu receptors are broadly divided into GRI and GRM. These receptors are distinguished based on structure, binding affinities, and signaling. On the left, the table illustrates GRI subunits and their corresponding coding genes. The proteins and coding genes belonging to the GRM families are depicted on the right panel.
Receptors 04 00004 g002
Figure 3. Membrane to nuclei signaling pathways resulting from the activation of Glu receptors in Radial Glia cells. This image depicts our current understanding of the translational and transcriptional modulation resulting from the activation of the Glu receptor subtypes present on the membrane of radial glial cells. GRIA (AMPAR): Leads to calcium influx, activating Ca2+/CaM (calmodulin), Protein Kinase C (PKC), and further downstream signaling. GRIN (NMDAR): Facilitates calcium influx, which contributes to activation of CAMKIII (Calcium/Calmodulin-Dependent Protein Kinase III) and MAPK (Mitogen-Activated Protein Kinase) signaling cascades. GRM 1,5 (Group I GRM): Gq-coupled receptors activate DAG (Diacylglycerol) and Ca2+/CaM pathways. GRM 2,3 (Group II GRM): Gi-coupled receptors inhibit downstream translation by reducing eEF2 activity. PKC activates transcriptional regulator YY1, influencing gene expression. MAPK/ERK Pathway: Both calcium and PKC pathways converge on MAPK, leading to ERK activation and regulation of nuclear transcription factors like CREB and FOS for gene expression. Translation Regulation: CAMKIII and eEF2K (eukaryotic elongation factor 2 kinase) modulate eEF2, while mTOR (mechanistic target of rapamycin) supports protein translation. ERK activates transcription factors, influencing the expression of target genes. This integration of signaling pathways highlights the roles of glutamate receptors in regulating gene expression and protein synthesis, essential for cellular adaptations and neuronal functions.
Figure 3. Membrane to nuclei signaling pathways resulting from the activation of Glu receptors in Radial Glia cells. This image depicts our current understanding of the translational and transcriptional modulation resulting from the activation of the Glu receptor subtypes present on the membrane of radial glial cells. GRIA (AMPAR): Leads to calcium influx, activating Ca2+/CaM (calmodulin), Protein Kinase C (PKC), and further downstream signaling. GRIN (NMDAR): Facilitates calcium influx, which contributes to activation of CAMKIII (Calcium/Calmodulin-Dependent Protein Kinase III) and MAPK (Mitogen-Activated Protein Kinase) signaling cascades. GRM 1,5 (Group I GRM): Gq-coupled receptors activate DAG (Diacylglycerol) and Ca2+/CaM pathways. GRM 2,3 (Group II GRM): Gi-coupled receptors inhibit downstream translation by reducing eEF2 activity. PKC activates transcriptional regulator YY1, influencing gene expression. MAPK/ERK Pathway: Both calcium and PKC pathways converge on MAPK, leading to ERK activation and regulation of nuclear transcription factors like CREB and FOS for gene expression. Translation Regulation: CAMKIII and eEF2K (eukaryotic elongation factor 2 kinase) modulate eEF2, while mTOR (mechanistic target of rapamycin) supports protein translation. ERK activates transcription factors, influencing the expression of target genes. This integration of signaling pathways highlights the roles of glutamate receptors in regulating gene expression and protein synthesis, essential for cellular adaptations and neuronal functions.
Receptors 04 00004 g003
Table 1. Expression of Glutamate Receptor subunits in Neuronal and Non-neuronal cells of the Retina. This table shows the expression of diverse Glu receptor subunits in different experimental models.
Table 1. Expression of Glutamate Receptor subunits in Neuronal and Non-neuronal cells of the Retina. This table shows the expression of diverse Glu receptor subunits in different experimental models.
Cell TypeIdentified Receptor Subtypes and SubunitsOrganismDetection TechniquesReferences
Horizontal CellsGRIN1, GRIA1, GRIA4, GRIK2Rat, mouse, rabbitImmunolabeling, scRNA sequencing[101,201,202]
Bipolar CellsGRIN1, GRIN2A, GRIN2B, GRIK1, GRM 4, GRM 6, GRM 7Rat, rabbitImmunofluorescence, western blot, immunocytochemistry.[27,167,203]
Amacrine CellsGRIN1, GRIN2B, GRIA1, GRIA4, GRIK2, GRM1, GRM2, GRM5, GRMR3Rat, cat, mouse, chick embryo, Goldfish Immunolabeling, immunocytochemistry, immunoelectron microscopy[164,167,204,205]
Retinal Ganglion CellsGRIA1, GRIA4, GRIK2, GRIN1, GRIN 2A, GRIN2B
GRMR1
Mouse, ratImmunolabeling, immunocytochemistry.[167,204,206]
Müller CellsGRIA4, GRIN1RatImmunocytochemistry, Western blot. [192,206,207,208]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Oyetayo, B.; Mendoza-Silva, Y.; Subair, T.; Hernández-Kelly, L.C.; Felder-Schmittbuhl, M.-P.; Olivares-Bañuelos, T.N.; Ortega, A. Glutamate Receptor Signaling in Retina Müller Cells: Plausible Role in Neurodegeneration. Receptors 2025, 4, 4. https://doi.org/10.3390/receptors4010004

AMA Style

Oyetayo B, Mendoza-Silva Y, Subair T, Hernández-Kelly LC, Felder-Schmittbuhl M-P, Olivares-Bañuelos TN, Ortega A. Glutamate Receptor Signaling in Retina Müller Cells: Plausible Role in Neurodegeneration. Receptors. 2025; 4(1):4. https://doi.org/10.3390/receptors4010004

Chicago/Turabian Style

Oyetayo, Bolaji, Yurixy Mendoza-Silva, Temitayo Subair, Luisa C Hernández-Kelly, Marie-Paule Felder-Schmittbuhl, Tatiana N. Olivares-Bañuelos, and Arturo Ortega. 2025. "Glutamate Receptor Signaling in Retina Müller Cells: Plausible Role in Neurodegeneration" Receptors 4, no. 1: 4. https://doi.org/10.3390/receptors4010004

APA Style

Oyetayo, B., Mendoza-Silva, Y., Subair, T., Hernández-Kelly, L. C., Felder-Schmittbuhl, M.-P., Olivares-Bañuelos, T. N., & Ortega, A. (2025). Glutamate Receptor Signaling in Retina Müller Cells: Plausible Role in Neurodegeneration. Receptors, 4(1), 4. https://doi.org/10.3390/receptors4010004

Article Metrics

Back to TopTop