Fact-checked by Grok 2 weeks ago

Kainate receptor

Kainate receptors are a subclass of ionotropic glutamate receptors (iGluRs) that function as tetrameric, ligand-gated cation channels mediating fast excitatory synaptic transmission in the central nervous system. Composed of five subunits (GluK1–GluK5), they assemble primarily as homomers or heteromers of GluK1–GluK3, while GluK4 and GluK5 require co-assembly with the former to form functional receptors; these subunits feature an N-terminal domain (NTD) for subunit assembly, a ligand-binding domain (LBD), and a transmembrane domain (TMD) forming the ion pore. Named after the agonist kainic acid, which selectively activates them over other iGluRs like AMPA receptors, kainate receptors exhibit unique biophysical properties, including slower desensitization and modulation by auxiliary proteins such as Neto1 and Neto2, which enhance trafficking and gating. These receptors play diverse roles both pre- and postsynaptically, modulating release (e.g., glutamate and ) at presynaptic sites to fine-tune synaptic transmission and influencing postsynaptic excitatory currents with intermediate kinetics compared to receptors. Extrasynaptic kainate receptors further regulate neuronal excitability and network oscillations, contributing to , activity, and circuit refinement; they also signal metabotropically via G-protein coupling, independent of , to affect long-term neuronal adaptations. Dysregulation of kainate receptors has been implicated in neurological disorders, including and mood disorders, highlighting their therapeutic potential. Pharmacologically, kainate receptors are activated by glutamate and selective agonists like kainate and SYM2081, which bind the bilobed LBD to induce conformational changes that open the cation-selective , allowing Na⁺ and K⁺ influx. Competitive antagonists such as NBQX and UBP310 block this binding, while positive allosteric modulators at the LBD dimer interface (e.g., influenced by ions like Na⁺ and Cl⁻) alter desensitization rates; at sites like Q/R in GluK2 subunits further tunes Ca²⁺ permeability and channel properties. Emerging therapies include gene therapies like AMT-260 targeting GluK2 for (as of 2024) and novel positive allosteric modulators. Advances in cryo-EM, including time-resolved studies as of 2024, have elucidated their gating mechanisms, revealing a dimer-of-dimers and stepwise pore opening that underpin their specialized functions.

Molecular Structure and Composition

Subunit Composition

Kainate receptors are tetrameric ion channels composed of five distinct subunits, known as GluK1 through GluK5, which are encoded by the GRIK1 to GRIK5 genes, respectively. These genes are located on different in the : GRIK1 on chromosome 21q21.2, GRIK2 on 6q16.3, GRIK3 on 1p34.3, GRIK4 on 11q23.3, and GRIK5 on 19q13.2. The subunits exhibit notable , particularly within the ligand-binding and transmembrane domains, where GluK1–GluK3 share approximately 75–80% identity, while overall similarity to GluK4 and GluK5 is lower at around 40%. This conservation underscores their shared architectural features despite functional differences. GluK1–GluK3 subunits are capable of forming functional homomeric channels when expressed alone, whereas GluK4 and GluK5 do not produce functional homomers and instead require co-assembly with GluK1–GluK3 subunits to form heteromeric receptors. Each subunit features a modular , including an amino-terminal (NTD) that varies across subtypes, influencing receptor assembly and . For instance, the NTDs of GluK1–GluK3 display 68–75% sequence identity among themselves, contributing to preferential heteromerization within this group. The C-terminal tails also differ significantly in length and composition; GluK2, for example, possesses a long intracellular rich in sites, such as serines 846 and 868, which regulate receptor trafficking and activity through kinases like PKC. These variations enable diverse regulatory mechanisms while maintaining the core tetrameric framework of kainate receptors.

Overall Architecture

Kainate receptors form tetrameric assemblies consisting of four subunits, typically homotetramers of GluK2 or heterotetramers involving other GluK subunits, each comprising an amino-terminal (ATD), ligand-binding domain (LBD), (TMD), and C-terminal domain. The overall quaternary structure exhibits a layered , with the extracellular ATD and LBD layers capping the membrane-spanning TMD that houses the ion-conducting pore. This modular architecture enables coordinated responses to glutamate binding, with the ATD layer adopting a dimer-of-dimers configuration that stabilizes the tetrameric interface through symmetric interactions between subunits. The ATD, located at the distal extracellular region, features a clamshell fold similar to bacterial periplasmic binding proteins and contributes to subunit assembly and allosteric modulation. Below it, the four LBDs form a compact layer where each domain undergoes a clamshell upon agonist binding, such as kainate or glutamate, which propagates conformational changes to the underlying TMD. The TMD consists of three transmembrane helices (, M3, M4) and a re-entrant M2 loop per subunit, collectively forming a central with pseudo-four-fold symmetry. Domain interactions, particularly at the ATD-LBD and LBD-TMD interfaces, ensure structural integrity and across the layers. The pore's selectivity is defined by the conserved SYTANLAAF within the M2 of each subunit, as exemplified in GluK2, which permits high permeability to Na⁺ and K⁺ ions while restricting Ca²⁺ influx (P_Ca/P_Na ≈ 0.1-0.5 depending on ). This non-selective cation conductance underlies the receptor's role in excitatory signaling. Recent cryo-EM s of GluK2 homotetramers in the ligand-free apo state have been resolved at 3.6 Å, illustrating the resting conformation with compact LBD dimers and a closed pore. Cryo-EM models from 2024-2025 further detail ligand-bound and desensitized states of GluK2 at resolutions of 3.4-4.3 , revealing LBD rearrangements where agonist-induced dimer separation in the desensitized state decouples the LBD layer from the TMD, leading to pore constriction without ATD disruption. These insights underscore the receptor's dynamic architecture, with the ATD maintaining tetrameric stability amid LBD flexibility.

Auxiliary Subunits

Kainate receptors associate with transmembrane auxiliary subunits known as neuropilin and tolloid-like (Neto) proteins, specifically Neto1 and Neto2, which co-assemble primarily with heteromeric complexes containing GluK2 and GluK3 subunits to modulate receptor properties.01085-4) These single-pass transmembrane proteins feature extracellular and receptor class A (LDLa) domains that interact with the receptor's amino-terminal domain (ATD) and ligand-binding domain (LBD), as well as intracellular C-terminal tails that influence assembly and trafficking. Neto1 and Neto2 exert distinct functional impacts on kainate receptor gating; Neto2 prominently slows the onset of desensitization and deactivation while accelerating recovery from desensitization, whereas Neto1 more effectively enhances potency by reducing the EC50 for kainate and promotes faster recovery from desensitization in certain subunit combinations. These modulations arise from Neto proteins stabilizing the receptor in activated states, with Neto2 showing a stronger effect on slowing desensitization kinetics (increasing τdes from ~5 ms to ~25 ms in GluK2 homomers). Recent cryo-EM structures of the GluK2-Neto2 complex reveal how Neto2 integrates structurally with the core tetrameric architecture of the receptor through multiple extracellular interfaces, including CUB1 binding to the ATD and CUB2/LDLa interactions with LBD loops and lower lobes, which stabilize LBD dimers by maintaining a fixed inter-dimer distance and preventing excessive LBD layer compression during . These interactions, spanning ~1,000 Ų at the alone, ensure Neto2's role in fine-tuning gating without altering the fundamental tetrameric pore formation. In contrast to their prominent role in AMPA receptors, where cornichon homolog (CNIH) proteins like CNIH-2/3 robustly modulate trafficking and gating, CNIHs exhibit only a minor association with kainate receptors, as evidenced by distinct interactomes that favor Neto proteins for kainate complexes.00128-3) Neto2 expression is particularly enriched in the and , where it co-localizes with kainate receptors in granule cells and pyramidal neurons, supporting region-specific modulation.

Biophysical Properties

Ion Selectivity and Conductance

Kainate receptors exhibit cation-selective ion permeability, primarily allowing influx of Na⁺ and efflux of K⁺ ions, with notably low permeability to Ca²⁺. The relative permeability to Ca²⁺ compared to monovalent cations (P_Ca/P_mono) is approximately 0.47 for RNA-edited forms of the GluK2 subunit (formerly GluR6), rendering Ca²⁺ influx minimal relative to that through NMDA receptors, which have P_Ca/P_Na ratios exceeding 5. This low Ca²⁺ permeability is determined by RNA editing at the Q/R site in the TM2 region of the pore-lining segment, where substitution of glutamine (Q) with arginine (R) reduces Ca²⁺ entry while unedited forms display higher ratios around 1.2. In heteromeric assemblies, such as those involving GluK1 and GluK2, the overall Ca²⁺ permeability remains subdued, typically below 1 relative to Na⁺, contributing to modest Ca²⁺ signaling compared to other ionotropic glutamate receptors. Single-channel conductance of kainate receptors varies with subunit composition but is generally in the range of 20–40 for homomeric GluK2 channels under physiological conditions. Patch-clamp recordings of recombinant GluK2 receptors reveal a weighted mean single-channel conductance of approximately 23 , with subconductance levels contributing to the overall current flow during activation. In heteromeric receptors, such as GluK2/GluK5, conductance can increase toward the upper end of this range due to altered pore architecture, enhancing total ionic flux without substantially altering selectivity. These conductance properties position kainate receptors as efficient mediators of depolarizing currents, though less so than NMDA receptors with their higher conductances. Kainate receptor currents display inward , arising from voltage-dependent block by endogenous intracellular s such as . This block is more pronounced at positive membrane potentials, reducing outward current and promoting inward flow of cations at resting potentials, with a rectification index often exceeding 3-fold. The involves polyamines binding within the channel pore, particularly in Ca²⁺-permeable variants, and is attenuated in heteromers or by auxiliary subunits like Neto proteins, which facilitate polyamine permeation and reduce rectification. Experimental patch-clamp studies confirm this voltage dependence, with intracellular spermine yielding a block affinity (K_d at 0 mV) of about 5.5 μM for kainate receptor subtypes. Patch-clamp electrophysiology further characterizes the open-state dynamics of kainate receptors, with mean open times typically ranging from 1 to 5 ms during application. For instance, single-channel recordings in response to kainate or related agonists like show mean open times of 2–3 ms for low-conductance states (~4 ), reflecting brief channel openings suited to fast synaptic . These durations can extend slightly with heteromer formation or positive , but remain shorter than those of NMDA receptors, emphasizing the role of kainate receptors in transient excitatory signaling.

Gating Mechanisms

The gating of kainate receptors (KARs) is initiated by the binding of glutamate to the ligand-binding domains (LBDs) of the receptor subunits, which induces a characteristic clamshell-like closure in each LBD. This closure, typically around 20°, separates the D2 lobes of the LBD and increases intersubunit distances, leading to an expansion of the LBD layer. These movements are transduced to the (TMD), where they pull on the pore-forming M3 helices, causing a coordinated kinking at conserved residues (e.g., L655 in GluK2) across all four subunits and widening the ion conduction pathway to permit cation influx. Dose-response relationships for KAR activation reflect the cooperative nature of this tetrameric , with half-maximal effective concentrations (EC50) for glutamate typically in the range of 100–500 μM, depending on subunit (e.g., ~159 μM for GluK2 homomers). The Hill coefficient for these responses often falls between 2 and 3, indicating positive arising from the requirement for multiple LBDs to bind for efficient opening, as opposed to independent subunit activation. Recent chimeric studies using systematic domain swaps between AMPA receptor subunit GluA2 and KAR subunit GluK2 have elucidated modular control of gating, highlighting distinct LBD-TMD coupling efficiencies between receptor subtypes. In these experiments, 14 chimeras revealed that the LBD and TMD form a tightly integrated "gating cassette," with GluK2's TMD showing reduced compatibility with GluA2's LBD, resulting in lower gating efficacy due to weaker allosteric transmission of conformational changes. Gating efficacy (ε) can be quantified as the ratio of peak open probability (Popen) to the fractional ligand occupancy, approximated by ε = Popen / ([L] / (Kd + [L])), where [L] is ligand concentration and Kd is the dissociation constant; this metric underscores how KAR chimeras exhibit ~50-fold slower recovery from desensitization compared to AMPA counterparts, emphasizing hierarchical allosteric interactions. The activation kinetics of KAR gating are temperature-sensitive, with a Q10 value of approximately 3 for the of channel opening, reflecting accelerated conformational rearrangements at physiological temperatures relative to recordings. This temperature dependence aligns with broader behavior, where higher temperatures enhance the speed of LBD closure and TMD dilation without altering equilibrium affinity.

Desensitization Dynamics

Kainate receptors, particularly homomeric assemblies of the GluK2 subunit, exhibit rapid desensitization upon prolonged exposure to glutamate, characterized by a (τ_des) of approximately 4–10 ms. This rapid entry into the desensitized state is slightly faster than that observed for many subtypes, which typically desensitize with τ_des values around 5–10 ms, enabling kainate receptors to briefly sustain ionic flux before rundown. Recovery from desensitization occurs more slowly, with a (τ_rec) of about 100–500 ms for GluK2, allowing receptors to re-enter responsive states after agonist removal but limiting sustained signaling during continuous glutamate presence. The auxiliary subunit Neto2 significantly modulates these dynamics, slowing the rate of desensitization onset by 2–3 fold (increasing τ_des) while accelerating recovery by a similar factor (decreasing τ_rec). This modulation enhances the overall charge transfer through , as Neto2 stabilizes partially open conformations and facilitates quicker return to baseline availability, a property confirmed through electrophysiological recordings of GluK2-Neto2 complexes. Recent structural studies using cryo-EM have elucidated the molecular basis of desensitization in GluK2 receptors. In the desensitized state, separation of the ligand-binding domain (LBD) dimers disrupts the tight inter-subunit interactions at the D1-D1 interface, uncoupling the LBD layer from the (TMD). This uncoupling involves large in-plane rotations of the LBD, twisting the LBD-TMD linkers and enforcing tight closure of the pore at the bundle-crossing region, as observed in shallow- and deep-desensitized conformations stabilized by targeted . These kinetics underpin the steady-state response of kainate receptors during prolonged application, where the steady-state (I_ss) approximates I_peak / (1 + τ_des / τ_rec), reflecting the balance between desensitization and rates. This relationship highlights how variations in τ_des and τ_rec dictate the residual conductance, with Neto2 modulation shifting I_ss closer to I_peak values.

Receptor Assembly and Localization

Homomeric and Heteromeric Assemblies

Kainate receptors (KARs) assemble as tetrameric complexes from five subunit types, denoted GluK1 through GluK5, with homomeric assemblies formed exclusively by GluK1–GluK3 subunits. These homomers consist of four identical subunits arranged in a 4:0 , creating a symmetric structure where each subunit contributes to the ligand-binding domain (LBD), transmembrane domains, and pore. For instance, GluK2 homotetramers exhibit high affinity for kainate, with an EC50 of 4.8 μM, enabling robust activation at physiological concentrations. This assembly allows for functional channels capable of and rapid gating, though their biophysical properties differ from heteromers due to uniform subunit . Heteromeric KARs, in contrast, incorporate combinations of GluK1–GluK3 with GluK4 or GluK5, commonly featuring GluK2/GluK5 pairs in a 2:2 , where two copies of each subunit form the tetramer. In these assemblies, the LBDs organize into two heterodimers, with GluK5 subunits positioned proximal to the channel pore, influencing overall receptor without high-affinity binding at their sites. GluK5 primarily modulates desensitization and recovery rates rather than directly gating the pore, as its low affinity (EC50 >1 mM for glutamate) limits independent activation. This configuration enhances receptor stability and alters conductance compared to homomers. Assembly of functional KARs is regulated by intracellular signals, particularly endoplasmic reticulum (ER) retention motifs in GluK4 and GluK5 subunits, which include dileucine-based sequences that prevent their homomeric trafficking to the plasma membrane. These signals ensure that GluK4/5 only reach the surface in heteromeric complexes with GluK1–GluK3, promoting obligatory partnering for mature receptor formation. Recent advances, such as 2025 studies on GluK2-selective RNA aptamers, have demonstrated their ability to potentiate GluK2 homomeric channels by binding selectively to GluK2, with an EC50 of ~210 nM.

Subcellular Distribution

Kainate receptors are trafficked to the plasma membrane and anchored at specific subcellular locations through distinct motifs in their subunit C-termini. These include PDZ-binding domains in subunits such as , , and , which interact directly with the PDZ1 domain of , a key scaffolding protein in the postsynaptic density. This interaction stabilizes assembled heteromeric receptors, like , at postsynaptic sites and promotes their synaptic clustering by linking them to the cytoskeletal framework. In hippocampal neurons, kainate receptors display a compartmentalized between synaptic and extrasynaptic regions, with diffusion barriers in the plasma membrane restricting lateral mobility and maintaining spatial segregation. Immunogold electron microscopy reveals that GluK2 subunits are primarily postsynaptic at mossy fiber-CA3 synapses, exhibiting clustered labeling with a major peak within 20–30 nm of the postsynaptic membrane, while a smaller fraction appears extrasynaptic. Similar postsynaptic enrichment is observed for GluK1, though with additional presynaptic localization, underscoring the role of subunit composition in dictating precise positioning. Activity-dependent regulation further shapes kainate receptor localization, with neuronal activity promoting their insertion into the plasma membrane via post-translational modifications. (PKC) phosphorylation at sites like Ser846 and Ser868 on GluK2 modulates trafficking through the secretory pathway and , influencing surface expression levels in an activity-dependent manner; for instance, stimulation triggers PKC activation, which can enhance delivery of receptors under certain conditions while also facilitating for dynamic control. Super-resolution imaging techniques, including structured illumination microscopy, have confirmed the clustered organization of kainate receptors at mossy fiber synapses, highlighting their nanoscale proximity to the synaptic cleft and association with transsynaptic proteins like C1ql2, which further refines their postsynaptic alignment.

Expression Patterns

Kainate receptor subunits exhibit distinct regional expression patterns in the , with high levels of GluK2 and GluK5 observed in the , particularly along CA3 mossy fibers, where intense immunolabeling has been reported in models. In the , GluK2 is prominently expressed in granule cells, contributing to local synaptic modulation. Conversely, expression of these subunits is generally low in the , as evidenced by weak hybridization signals for GluK2 mRNA in cortical layers. Developmentally, kainate receptor subunit expression in peaks during the second postnatal week, with mRNA levels for most subunits rising from low birth levels to a maximum around postnatal day 14 before stabilizing or declining. In humans, adult expression data from the GTEx portal indicate enrichment of kainate receptor genes, such as GRIK1 (GluK1), in the compared to other brain regions. Subunit-specific patterns include transient high expression of GluK1 mRNA in the embryonic and early postnatal , peaking around birth in sensory areas before downregulation. GluK3 expression is notable in thalamic relay nuclei during , supporting relay neuron function. In disease contexts, kainate receptor subunits show downregulation in models of , with studies reporting 30–50% reductions in GluK2 expression in affected hippocampal and tissues, correlating with chronic activity. These changes highlight altered expression as a potential contributor to epileptogenic networks.

Physiological Functions

Presynaptic Modulation

Kainate receptors (KARs) located at presynaptic terminals exert bidirectional control over neurotransmitter release, primarily through metabotropic signaling pathways independent of their ionotropic function. Activation of presynaptic KARs inhibits release from in the and other regions by coupling to G-protein signaling, which suppresses inhibitory transmission and enhances overall network excitability. In contrast, at mossy fiber terminals in the CA3 region of the , low concentrations of kainate facilitate glutamate release via a similar G-protein-mediated mechanism, increasing the probability of presynaptic vesicle fusion without requiring calcium influx through the receptor channel itself. Heteromeric assemblies of GluK1 and GluK2 subunits predominate in presynaptic , forming the core structure responsible for these modulatory effects at mossy fiber synapses and terminals. These GluK1/GluK2 heteromers are enriched at presynaptic sites, where they integrate metabotropic signals to fine-tune release dynamics, distinguishing them from postsynaptic or extrasynaptic receptor populations. Physiologically, presynaptic KAR contributes to facilitation at mossy fiber-CA3 synapses, enhancing excitatory during high-frequency bursts in the 10–20 Hz range, which supports rapid processing in hippocampal . In the , KARs play a critical role in developmental synaptic refinement, as demonstrated by a 2024 study showing that their modulates climbing fiber onto Purkinje cells, ensuring proper elimination and circuit maturation. Desensitization of these receptors can limit prolonged presynaptic effects during sustained exposure.

Postsynaptic Signaling

Kainate receptor-mediated excitatory postsynaptic currents (EPSCs) display characteristically slow , with rise times of approximately 1–2 ms and times ranging from 10–50 ms, owing to the receptors' propensity for rapid desensitization upon glutamate binding. This desensitization prolongs the postsynaptic response beyond the brief glutamate transient in the synaptic cleft, enabling temporal summation of successive inputs and the emergence of sustained depolarizations termed plateau potentials, which can outlast individual synaptic events by hundreds of milliseconds. These properties distinguish kainate EPSCs from faster receptor-mediated currents and allow kainate receptors to integrate excitatory signals over longer timescales in specific neuronal populations. Despite their lower peak amplitudes, kainate EPSCs achieve significant postsynaptic amplification through their extended duration; the single-channel conductance of kainate receptors is roughly 5–10 fold smaller than that of receptors, yet the prolonged decay compensates by increasing overall charge transfer, often matching or exceeding that of AMPA-mediated events in terms of integrative impact. For context, kainate receptor conductances typically range from 5–20 per channel, contributing to smaller instantaneous currents but enabling a broader temporal window for . This amplification is particularly evident during repetitive stimulation, where enhances neuronal excitability without requiring higher receptor density. In hippocampal circuits, postsynaptic kainate receptors play a specialized role in , where they generate rhythmic bursting patterns that underpin network oscillations. Activation of GluK2/3-containing kainate receptors in CA1 , for instance, drives phase-locked firing to rhythms (4–8 Hz), promoting synchronized inhibitory output that gates principal cell activity and supports memory-related processes. This bursting arises from the slow EPSC , which facilitate burst initiation and maintenance through persistent sodium current interplay. Auxiliary proteins such as Neto1 further refine postsynaptic kainate receptor signaling in the CA1 region by enhancing efficacy in . Neto1 co-assembly slows desensitization, accelerates rise times to ~2.7 ms, and extends decay to ~70 ms while boosting current amplitude, thereby increasing excitatory drive and recruitment of network inhibition. In Neto1-deficient conditions, these enhancements are lost, underscoring its role in optimizing synaptic integration for circuit stability.

Extrasynaptic Roles

Kainate receptors () located outside synaptic junctions, particularly those containing the GluK2 subunit, can be activated by ambient glutamate through volume transmission, allowing the to diffuse and influence neuronal excitability across broader networks. This mechanism enables low concentrations of glutamate to engage perisynaptic and extrasynaptic GluK2-containing on hilar mossy cells in the , triggering inward currents and robust neuronal firing without reliance on synaptic release. Such activation by blocking glutamate transporters, which elevates extracellular glutamate levels, underscores the sensitivity of these extrasynaptic receptors to glutamate signaling, thereby modulating hippocampal circuit activity and potentially contributing to network synchronization. Low-level activation of extrasynaptic promotes by inducing rapid desensitization, which limits excessive ion influx and mitigates glutamate-induced during pathological conditions like ischemia. The profound desensitization kinetics of , with time constants around 4-5 ms, serve as an intrinsic safeguard, reducing the risk of neuronal overexcitation by confining receptor responses to brief pulses of . For instance, the selective KAR SYM 2081 elicits desensitization-dominant responses that preserve neuronal viability in hypoxic-ischemic models, downregulating excitotoxic GluK1 expression while stabilizing other subunits to maintain balanced signaling. KARs are expressed in astrocytes, particularly upregulated following seizure activity, where they contribute to gliotransmission by sensing extracellular glutamate and modulating calcium-dependent release of signaling molecules. In hippocampal astrocytes, subunits such as GluK1 and GluK2 appear post-status epilepticus, potentially enhancing astrocyte reactivity and glutamate release that influences nearby neuronal networks. This expression allows astrocytes to integrate ambient glutamate signals via KARs, facilitating Ca²⁺ waves that drive gliotransmitter output, such as glutamate itself, to fine-tune inhibitory transmission in interneurons. Emerging research highlights the involvement of extrasynaptic GluK1-containing in pathways, where they contribute to central nociceptive in the trigeminocervical system. Antagonists targeting GluK1, like LY466195, suppress trigeminocervical complex activity and dural in preclinical models, suggesting extrasynaptic GluK1 amplifies signaling through tonic glutamate modulation. Recent structural advances, including 2024 cryo-EM studies, have elucidated KAR gating mechanisms in extrasynaptic contexts, revealing a dimer-of-dimers arrangement that underpins their roles in network oscillations and excitability.

Involvement in Synaptic Plasticity

Short-Term Plasticity

Kainate receptors play a key role in short-term , particularly through presynaptic mechanisms that modulate glutamate release probability in response to high-frequency . At hippocampal mossy fiber-CA3 synapses, of presynaptic kainate receptors (GluKs) during repetitive activity enhances the initial low release probability (P_r), promoting frequency-dependent facilitation and counteracting potential short-term . This process involves ionotropic influx of cations, which triggers rapid intracellular signaling to increase vesicular release, thereby amplifying synaptic strength on a timescale of seconds. A representative example is observed at mossy fiber-CA3 synapses, where low concentrations of kainate (20–50 nM) relieve short-term depression induced by sustained activity, resulting in a 20–50% increase in excitatory postsynaptic currents (EPSCs). This facilitatory effect is most prominent during trains of stimuli, where kainate receptor activation boosts successive responses, enhancing overall synaptic output without altering baseline transmission. Such modulation underscores the receptors' role in dynamic adjustment of network activity during physiological patterns like theta rhythms. The kinetics of kainate receptor-mediated facilitation are rapid, aligning with the receptors' localization on presynaptic terminals, enabling them to respond to spillover glutamate from nearby synapses during high-frequency bursts. Recovery from these modulatory effects occurs over minutes, preventing prolonged alterations. GluK2-containing heteromers are critical for this presynaptic facilitation, as demonstrated by genetic studies showing markedly reduced paired-pulse and facilitation in GluK2 mice compared to wild-type controls. These heteromers, likely incorporating low-affinity subunits, confer specificity to the facilitatory response at mossy fiber terminals, distinguishing it from other synaptic loci.

Long-Term Potentiation and Depression

Kainate receptors (), particularly those containing the GluK2 subunit, play a crucial role in the induction of (LTP) at hippocampal mossy fiber-CA3 synapses. Presynaptic GluK2 KARs facilitate glutamate release in a frequency-dependent manner, which is essential for triggering the presynaptic mechanisms underlying LTP. This form of LTP is induced by high-frequency tetanic at approximately 100 Hz, typically consisting of multiple 1-second trains separated by 10 seconds, leading to a 2- to 3-fold increase in synaptic efficacy that lasts for hours or longer. Long-term depression (LTD) at mossy fiber-CA3 synapses involves KAR-mediated postsynaptic through slow excitatory postsynaptic currents (EPSCs), which activate endocannabinoid synthesis and retrograde signaling to suppress presynaptic glutamate release. These slow EPSCs, lasting hundreds of milliseconds, provide the sustained necessary to mobilize endocannabinoids like , which act on presynaptic CB1 receptors to induce LTD following low-frequency stimulation (1 Hz). This mechanism ensures activity-dependent weakening of synaptic transmission, contributing to the bidirectional control of synaptic strength by KARs. In the cerebellum, GluK2 KARs contribute to LTD at climbing fiber-Purkinje cell , where their activation regulates synapse elimination during developmental refinement and adult . A 2024 study demonstrated that GluK2 knockout impairs climbing fiber-Purkinje LTD, leading to persistent multi-innervation and disrupted , highlighting KARs' role in maintaining synaptic integrity through LTD-dependent pruning. Pharmacological blockade of with antagonists such as LY382884 not only prevents mossy LTP but also disrupts metaplasticity by eliminating the prior activity-dependent modulation of LTP thresholds. For instance, on mossy heterosynaptically lowers the threshold for LTP at nearby synapses, an associative metaplastic effect that is abolished by KAR antagonists, thereby altering the capacity for subsequent .

Developmental Regulation

Kainate receptors (), particularly those incorporating the GluK2 and GluK5 subunits, exhibit a distinct temporal expression profile during , with upregulation occurring in the early postnatal period to support refinement. In , mRNA for GluK2 and GluK5 is detectable during embryonic stages but peaks during the late embryonic to early postnatal phases (P0-P7), coinciding with intense and network maturation. This high expression facilitates the refinement of synapses by modulating presynaptic glutamate release and synaptic efficacy. Following this peak, expression levels of these subunits decline post-P10 and into adulthood, shifting KAR function from prominent developmental roles to more subtle modulatory influences in mature circuits. KAR activation plays a crucial role in , particularly by promoting the maturation of dendritic spines in the . The GluK2 subunit interacts directly with the potassium-chloride cotransporter KCC2, stabilizing spine structures and enhancing their morphological development in hippocampal neurons. This interaction is essential for the transition from immature filopodia-like protrusions to mature mushroom-shaped spines, thereby supporting the formation of functional excitatory synapses during early postnatal development. Endogenous activation of these receptors regulates network activity and glutamate release, further aiding the structural and functional refinement of hippocampal circuits. During critical periods of sensory development, KARs contribute to the formation of topographic maps, such as in the . In this region, KARs modulate at thalamocortical synapses onto layer IV granule cells, ensuring precise wiring and map stabilization during the first two postnatal weeks. Disruption of KAR function during this window impairs afferent segregation and sensory map organization. Recent sequencing studies have revealed epigenetic mechanisms underlying GRIK during neural development, including dynamic patterns that influence KAR subunit expression. For instance, single-cell profiling has identified bivalent states involving H3K4me1 and at the Grik1 locus, which poise the for timely activation in differentiating neurons. These findings highlight how epigenetic modifications, such as , fine-tune KAR expression to align with developmental milestones in excitatory circuit assembly.

Pharmacology and Therapeutics

Agonists and Endogenous Ligands

The primary endogenous ligand for kainate receptors is glutamate, which activates these ionotropic glutamate receptors with an EC50 of approximately 500 μM across various recombinant and native preparations. Quisqualate, a synthetic structurally related to glutamate, exhibits weak potency at kainate receptors, with an EC50 around 3 mM for homomeric GluK1 (GluR5) receptors, rendering it far less effective than other ligands. Among synthetic agonists, serves as a prototypical and highly selective activator of kainate receptors, displaying an EC50 of about 50 μM and characteristically producing non-desensitizing or slowly desensitizing responses compared to glutamate, which facilitates its use in electrophysiological studies of receptor function. α-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid () acts as a partial agonist at kainate receptors with low efficacy and an EC50 of roughly 500 μM, activating the channel but eliciting smaller currents than full agonists like . Domoic acid, a neurotoxic analog of kainic acid derived from marine algae, demonstrates high potency and subunit selectivity, particularly preferring homomeric GluK1 receptors with an EC50 of 1 μM, while maintaining lower micromolar affinity for other low-affinity kainate subunits like GluK2 and GluK3.

Antagonists and Negative Modulators

Kainate receptors (), composed of GluK1–5 subunits, are modulated by a variety of antagonists that inhibit receptor activation through competitive or non-competitive mechanisms, thereby reducing excitatory neurotransmission in the . Competitive antagonists bind to the , preventing access, while non-competitive antagonists and negative allosteric modulators interfere with channel gating or desensitization via distinct sites. These compounds have been instrumental in dissecting KAR functions and exploring therapeutic potential in conditions like and . Among competitive antagonists, 6-cyano-7-nitroquinoxaline-2,3-dione (CNQX) is a widely used non-selective that blocks both and kainate receptors by competing with glutamate at the ligand-binding site, with an of approximately 1.5 μM for kainate-evoked currents and 0.3 μM for receptors. This lack of selectivity limits its use for KAR-specific studies but has facilitated early identification of KAR-mediated responses in synaptic transmission. In contrast, 2,3-dioxo-6-nitro-1,2,3,4-tetrahydrobenzoquinoxaline-7-sulfonamide (NBQX) exhibits greater potency at receptors ( ~0.15 μM) but still antagonizes GluK2-containing KARs with an of about 2–4.8 μM, offering improved selectivity over earlier quinoxalinediones while displaying GluK2 preference in heteromeric assemblies. Non-competitive antagonists, such as talampanel (GYKI 53773), exert use-dependent blockade by binding within the channel pore of /kainate receptors, stabilizing a closed state and inhibiting flux with higher during prolonged ; it shows moderate potency at (IC50 ~10–30 μM) alongside stronger AMPA inhibition. This mechanism reduces without fully occluding the orthosteric site, contributing to its evaluation in models where KAR hyperactivity drives seizures. Negative allosteric modulators like LY382884 target presynaptic GluK1 (GluR5)-containing with high selectivity (>100-fold over AMPA receptors), binding to an allosteric site to suppress glutamate release; it inhibits kainate-induced currents with an IC50 of ~1 μM and has demonstrated in attenuating behaviors in preclinical studies. Recent structural investigations, including 2025 cryo-EM studies of chimeric KAR constructs, have revealed subunit-specific allosteric pockets that enable design of more selective antagonists, such as those targeting GluK2/GluK5 interfaces to enhance potency and minimize off-target effects on receptors. These advances underscore the potential for precision in modulating KARs for therapeutic applications.

Positive Allosteric Modulators and Emerging Therapies

Positive allosteric modulators (s) of kainate receptors bind to sites distinct from the orthosteric -binding domain, enhancing receptor activation by increasing or slowing desensitization, thereby amplifying excitatory signaling without directly gating the . Cyclothiazide, a benzothiadiazine derivative, exemplifies an early PAM that primarily targets receptors but also modulates kainate receptors by reducing desensitization rates. This action prolongs open times, potentially influencing synaptic transmission where kainate receptors contribute to presynaptic facilitation. Recent advances have identified heterocyclic compounds as subtype-selective PAMs, particularly for GluK2-containing receptors complexed with Neto2 auxiliary subunits, which are critical for receptor trafficking and gating . A 2025 comprehensive review highlights these heterocycles, such as modified benzothiadiazine dioxides, which enhance GluK2-Neto2 currents by stabilizing the open state and shifting EC50 values, offering improved selectivity over traditional non-subtype-specific modulators. These developments address limitations in earlier PAMs by minimizing off-target effects on receptors, paving the way for targeted interventions in disorders involving dysregulated kainate signaling. As of 2025, structural studies using cryo-EM have further elucidated allosteric sites, supporting design of selective modulators. Emerging RNA-based tools, such as , represent innovative PAMs with high specificity. A 2025 study in Nature Scientific Reports describes the GluK2-selective aptamer U9, which acts as a potentiator by an allosteric , resulting in a approximately 2-fold increase in and an EC50 of 210 nM, without altering desensitization rates. This subunit selectivity distinguishes U9 from broad-spectrum modulators, enabling precise enhancement of GluK2 homomers or heteromers while sparing other ionotropic glutamate receptors. Therapeutically, kainate receptor modulation holds promise across neurological conditions. Antagonists, including analogs of —a non-competitive negative effective against both and kainate receptors—have demonstrated antiseizure activity in models by suppressing excessive excitation, with itself approved for refractory partial-onset seizures. In contrast, PAMs exhibit potential for mood disorders; preclinical data suggest they enhance and antidepressant-like effects in models by boosting kainate-mediated signaling in mood-regulating circuits. Clinical translation is advancing, particularly for antagonists in . A Phase II trial of BGG492, an /kainate blocker, for acute treatment did not meet its primary endpoint for headache (p > 0.05 vs. ), though it showed comparable pain-free responses to in some measures. Similarly, tezampanel, a competitive kainate , achieved significant in acute settings during Phase II studies, underscoring the therapeutic relevance of targeting kainate receptors to mitigate glutamate-driven . These efforts highlight emerging therapies that leverage allosteric for improved precision and reduced side effects in CNS disorders. No kainate receptor-specific drugs are clinically approved as of November 2025.

References

  1. [1]
    Structure, Function, and Pharmacology of Glutamate Receptor Ion ...
    Kainate receptor subunits GluK1-3 can assemble as functional homomers and heteromers, but GluK4-5 must coexpress with GluK1-3 to form functional receptors. NMDA ...
  2. [2]
    Exciting Times: New Advances Towards Understanding the ...
    Kainate receptors (KARs) are glutamate-gated ion channels that play fundamental roles in regulating neuronal excitability and network function in the brain.
  3. [3]
    Structural and compositional diversity in the kainate receptor family
    The kainate receptors (KARs) are members of the ionotropic glutamate receptor family and assemble into tetramers from a pool of five subunit types (GluK1–5).
  4. [4]
    Entry - GLUTAMATE RECEPTOR, IONOTROPIC, KAINATE 1; GRIK1
    Genetic and physical mapping of the GLUR5 glutamate receptor gene on human chromosome 21. Hum. Genet. 94: 565-570, 1994. [PubMed: 7959697, related citations] ...
  5. [5]
  6. [6]
  7. [7]
  8. [8]
  9. [9]
    A pharmacological profile of the high-affinity GluK5 kainate receptor
    Oct 5, 2016 · ... GluK1-5, of similar size (908–980 aa). GluK1-3 share 75–80% amino acid sequence identity while the similarity to GluK4-5 is only 40% and the ...
  10. [10]
    Mapping the Ligand Binding Sites of Kainate Receptors - NIH
    GluK1–3 are low affinity kainate binding subunits, whereas GluK4/5 bind kainate with high affinity. A number of these subunits can be present in any given cell ...
  11. [11]
    Preferential assembly of heteromeric kainate and AMPA receptor ...
    Oct 23, 2017 · Within each family, there are four AMPA receptor subunits, five kainate receptor subunits, seven NMDA receptor subunits, and two delta receptor ...Missing: variations | Show results with:variations
  12. [12]
    Modulation of GluK2a Subunit-containing Kainate Receptors by 14 ...
    Additionally, we determined that phosphorylation of several serine residues at the GluK2a C-terminal tail is required for this protein/protein interaction.
  13. [13]
    Amino-terminal domains of kainate receptors determine the ... - PNAS
    The kainate receptor (KAR) is a subfamily of glutamate receptors that mediates excitatory synaptic transmission in the central nervous system.
  14. [14]
    Kainate receptor channel opening and gating mechanism - Nature
    May 22, 2024 · Kainate receptors, a subclass of ionotropic glutamate receptors, are tetrameric ligand-gated ion channels that mediate excitatory neurotransmission.<|control11|><|separator|>
  15. [15]
    Architecture and structural dynamics of the heteromeric GluK2/K5 ...
    Mar 16, 2021 · Kainate receptors (KARs) are L-glutamate-gated ion channels that regulate synaptic transmission and modulate neuronal circuits.
  16. [16]
    Domain organization and function in GluK2 subtype kainate receptors
    Apr 19, 2010 · iGluRs are tetrameric assemblies in which the extracellular amino terminal domain (ATD) and ligand-binding domain (LBD) both assemble as a dimer of dimers.Missing: SYTAN | Show results with:SYTAN
  17. [17]
    Kainate induces an intracellular Na+-activated K+ current in cultured ...
    ... receptor-channels, which are highly permeable to Na+ and K+ and less permeable to Ca2+ ions (Hatt et al. 1988; Colquhoun et al. 1992). When activated, these ...
  18. [18]
  19. [19]
    Structure and gating of kainate receptors - Frontiers
    The KAR family of iGluRs comprises five subunits, GluK1-GluK5, which are encoded by the GRIK1-GRIK5 genes. KARs are widely expressed throughout CNS, with ...
  20. [20]
    Neto1 and Neto2: auxiliary subunits that determine key properties of ...
    Neto1 and Neto2 are auxiliary subunits of kainate receptors that determine their slow kinetics and high affinity for agonist. They are type-1 transmembrane ...
  21. [21]
    Activation of kainate receptor GluK2–Neto2 complex - Nature
    Aug 22, 2025 · During cryo-EM data processing, we identified three distinct populations of particles that yielded 3D reconstructions of GluK2 tetramer with ...
  22. [22]
    Neto2 Modulation of Kainate Receptors with Different Subunit ...
    Jun 1, 2011 · We recently identified Neuropilin and Tolloid like-2 (Neto2) as a novel accessory subunit of kainate receptors and showed that Neto2 modulates ...
  23. [23]
  24. [24]
    Determinants of Ca2+ permeability in both TM1 and TM2 ... - PubMed
    ... low Ca2+ permeability, whereas an R determines a higher Ca2+ permeability if ... Thus Ca2+ permeability of kainate receptor channels can vary, depending on ...
  25. [25]
    [PDF] Kainate receptor pore-forming and auxiliary subunits regulate ...
    Dec 18, 2015 · Kainate receptor heteromerization and auxiliary proteins attenuate polyamine block by enhancing blocker permeation, allowing unblock at more ...
  26. [26]
    Inward rectification of both AMPA and kainate subtype glutamate ...
    Article. Inward rectification of both AMPA and kainate subtype glutamate receptors generated by polyamine-mediated ion channel block.
  27. [27]
    Kainate receptor pore‐forming and auxiliary subunits regulate ...
    Dec 18, 2015 · The original protein structure is shown in white; the selectivity filter is highlighted in magenta. The Pro-containing α-helices (cyan, ...Abstract · Methods · Results · DiscussionMissing: SYTAN | Show results with:SYTAN
  28. [28]
    Kainate activated single channel cnrrents as revealed by domoic acid
    Small conductance cationic-selective channels of ≈4 pS and a mean open time of 2 to 3 ms were detected using both methods. Article PDF. Download to read the ...
  29. [29]
    Developmental Changes in AMPA and Kainate Receptor-Mediated ...
    May 25, 2005 · These findings provide the first detailed comparison of the quantal and single-channel properties of AMPA and kainate receptor-mediated ...
  30. [30]
    [PDF] Kainate-Receptor Assay
    Table 1: Glutamate Dependence. Receptor composition. EC50. Hill coefficient. GluK2 homomer. 159.32 ± 7.32 µM. 3.58. GluK2 / GluK5 heteromer.
  31. [31]
    Channel-Opening Kinetic Mechanism of Wild-Type GluK1 Kainate ...
    Kainate receptor channels are involved in regulating both excitatory and inhibitory neurotransmission. ... ms, from the best fit of the open-time distribution.
  32. [32]
    Modular control of AMPA and Kainate receptor gating revealed by ...
    Oct 14, 2025 · To address this, we created a comprehensive library of 14 reciprocal chimeric receptors between the AMPA receptor GluA2 and the kainate receptor ...Missing: SYTAN | Show results with:SYTAN<|control11|><|separator|>
  33. [33]
    Incomplete removal of extracellular glutamate controls synaptic ...
    Feb 22, 2021 · We chose to simply apply a Q10 (temperature coefficient) of 2.4 for all rates in the kinetic model for the following reasons: Multiplication by ...
  34. [34]
    Structural Insights into Kainate Receptor Desensitization - bioRxiv
    Structural Insights into Kainate Receptor Desensitization. Changping Zhou, View ... Posted April 23, 2025. Download PDF. Print/Save Options. Download PDF ...
  35. [35]
    A Molecular Determinant for Submillisecond Desensitization in ...
    Native AMPA receptors show desensitization time constants of 1 to about 10 milliseconds, but the underlying molecular determinants of these large differences ...
  36. [36]
    Article Measurements of the Timescale and Conformational Space ...
    Jul 7, 2020 · The time constant of recovery after trapping in resting conditions was 380 ± 150 ms, but for resting + CTZ, the recovery was much faster (30 ...
  37. [37]
    Neto2 modulation of kainate receptors with different subunit ...
    Jun 1, 2011 · In addition, Neto2 markedly increased the rate at which some receptors recovered from desensitization. These results suggest that Neto2 ...
  38. [38]
    [PDF] 1 Structural Insights into Kainate Receptor Desensitization ... - bioRxiv
    Apr 23, 2025 · Structural Insights into Kainate Receptor Desensitization. 1 ... this version posted April 23, 2025. ; https://doi.org/10.1101/2025.03 ...
  39. [39]
    Contributions of different kainate receptor subunits to the properties ...
    Oct 10, 2015 · The kainate EC50 was shifted to that of the GluK2 homomers (average EC50 = 3.5 ± 0.5 μM, n=4, p≤0.01 compared to GluK2/K5, p>0.5 compared to ...Missing: homotetramer | Show results with:homotetramer
  40. [40]
    Assembly Stoichiometry of the GluK2/GluK5 Kainate Receptor ...
    Mar 29, 2012 · Ionotropic glutamate receptors assemble as homo- or heterotetramers. One well-studied heteromeric complex is formed by the kainate receptor ...
  41. [41]
    Modulation of homomeric and heteromeric kainate receptors by the ...
    While GluK1–GluK3 subunits can form homomeric receptors, the GluK4 or GluK5 subunits only function in heteromeric assemblies with GluK1–GluK3 subunits.
  42. [42]
    GluK2 kainate receptor subunit-selective, potentiating RNA aptamer
    Sep 11, 2025 · The KA-2 subunit of excitatory amino acid receptors shows widespread expression in brain and forms ion channels with distantly related subunits.
  43. [43]
  44. [44]
  45. [45]
  46. [46]
  47. [47]
    A comparative analysis of kainate receptor GluK2 and GluK5 ...
    May 7, 2021 · Western blotting and immunohistochemistry revealed that a strong expression of GluK2 in the cortex, hippocampus, and cerebellum. These ...<|control11|><|separator|>
  48. [48]
    Kainate Receptors Modulating Glutamate Release in the Cerebellum
    Of these subunits, GluK1 and GluK3 may form homomeric or heteromeric functional receptors, while GluK4 and GluK5 may only participate in functional receptors ...Biogenic Amines In... · 1. Introduction · 1.1 Kainate Receptors
  49. [49]
    Kainate Receptor Gene Expression in the Developing Rat Brain
    13). The hybridization signal for GluR-6 in the cortex is generally low; but in the cingulate cortex this subunit is very prominent, ... receptor: a low affinity ...
  50. [50]
    The expression and binding of kainate receptors is modified in ...
    Additionally, kainate receptors in the rat have a low expression at birth, increasing its expression at the end of the first postnatal week, peaking at ...
  51. [51]
    Kainate receptor gene expression in the developing rat brain
    Sep 1, 1994 · The GluR-5 gene shows peaks of expression around the period of birth in the sensory cortex (layers II, III, and IV), in CA1 hippocampal ...
  52. [52]
    Kainate receptors in the developing neuronal networks - ScienceDirect
    Sep 1, 2021 · In the adult stage, subunits GluK2 and GluK5 are abundantly expressed in various areas of the brain, while the subunits GluK1, GluK3 and GluK4 ...Kainate Receptors In The... · 4. Morphogenic And... · 4.2. Kars And Maturation Of...
  53. [53]
    The Role of Kainate Receptors in the Pathophysiology of Hypoxia ...
    May 4, 2018 · Alterations in KAR subunit expression have been reported in both animal models of epilepsy and in clinical studies of human temporal lobe ...
  54. [54]
    Kainate Receptors: Role in Epilepsy - Frontiers
    Jun 21, 2018 · Aberrant glutamate receptor 5 expression in temporal lobe epilepsy lesions. Brain Res. 1311, 166–174. doi: 10.1016/j.brainres.2009.11.024.
  55. [55]
    Non-canonical Mechanisms of Presynaptic Kainate Receptors ...
    In the present review, we revisit the mechanisms underlying the non-canonical modus operandi of KARs effecting the bimodal control of glutamatergic transmission ...Abstract · Introduction · Hippocampus · Future Directions of...
  56. [56]
    Metabotropic actions of kainate receptors modulating glutamate ...
    Oct 1, 2021 · In general, presynaptic KARs modulate glutamate release through their metabotropic actions in a biphasic manner, with low KA concentrations ...
  57. [57]
    Role of GluK1 Kainate Receptors in Seizures, Epileptic Discharges ...
    Apr 23, 2014 · KARs occur predominantly as homomeric or heteromeric combinations of GluK1 and GluK2 or as heteromers of these subunits with GluK4 or GluK5 ...
  58. [58]
  59. [59]
    Protein kinase signalling requirements for metabotropic action ... - NIH
    The physiological correlate of this action – activation of PKC by kainate receptors – requires Gαi/o proteins. The cAMP/PKA system is well documented to ...Missing: signaling | Show results with:signaling
  60. [60]
  61. [61]
  62. [62]
  63. [63]
  64. [64]
    Kainate Receptor–Mediated Inhibition of Glutamate Release ...
    ... (20–50 nM) KA concentrations produce an increase in glutamate release in the mossy fiber–CA3 region (Contractor et al. 2001; Lauri et al. 2001a,b; Rodríguez ...
  65. [65]
  66. [66]
    Review Kainate Receptors in Health and Disease - ScienceDirect.com
    Oct 16, 2013 · (A) In the mossy fiber to CA3 synapses, activation of KARs is responsible of part of the frequency facilitation, such that the inclusion of a ...
  67. [67]
    Differential expression of kainate receptors in the basal ganglia of ...
    Sep 12, 1997 · During postnatal development, expression of the kainate receptor subunits was characteristically highest on postnatal day 1 and declined to ...Missing: profile | Show results with:profile
  68. [68]
    The Kainate Receptor Subunit GluK2 Interacts With KCC2 ... - Frontiers
    Aug 25, 2020 · Our results demonstrate that GluK2-KCC2 interaction plays an important role in the structural maturation of dendritic spines.
  69. [69]
    The Kainate Receptor Subunit GluK2 Interacts With KCC2 to ... - NIH
    Aug 26, 2020 · Our results demonstrate that GluK2-KCC2 interaction plays an important role in the structural maturation of dendritic spines.
  70. [70]
    Activation of kainate receptors controls the number of functional ...
    Activation of kainate receptors controls the number of functional glutamatergic synapses in the area CA1 of rat hippocampus - PMC.Missing: Q10 | Show results with:Q10
  71. [71]
    Development and Critical Period Plasticity of the Barrel Cortex - PMC
    The rodent barrel cortex offers an invaluable model system to investigate mechanisms underlying the formation of topographic maps and their plasticity during ...
  72. [72]
    Developmental Synaptic Plasticity at the Thalamocortical Input to ...
    Here we describe these studies that point to a key role for NMDA receptor-dependent synaptic plasticity, a role for kainate receptors and for a rapid maturation ...
  73. [73]
    Developmental synaptic plasticity at the thalamocortical input to ...
    Sensory information from the whiskers passes via the brain stem and thalamus to layer IV neurons in “barrel cortex” (Fig. 1A). Anatomical (Woolsey and Van der ...
  74. [74]
    Single-cell profiling of H3K4me1-H3K27me3 revealed bivalent ...
    Sep 1, 2025 · Grik1 encodes Glutamate Ionotropic Receptor Kainate Type Subunit 1, which is a ligand-gated ion channel that play a key role in excitatory ...
  75. [75]
  76. [76]
    Activation and Desensitization of Hippocampal Kainate Receptors
    Apr 15, 1997 · Most studies of kainate receptors report little or no change in agonist EC50 ... (1983) Cellular and synaptic basis of kainic acid-induced ...Missing: synthetic | Show results with:synthetic
  77. [77]
    Kainate Receptor Antagonists: Recent Advances and Therapeutic ...
    We reviewed recent advances in the field of competitive and non-competitive kainate receptor antagonists and their potential therapeutic applications.
  78. [78]
    CNQX | GluR antagonist | Mechanism - Selleck Chemicals
    CNQX (6-cyano-7-nitroquinoxaline-2,3-dione) is an AMPA and kainate receptor antagonist with IC50 values are 0.3 μM and 1.5 μM for AMPA and kainate receptors, ...<|separator|>
  79. [79]
    NBQX | AMPA Receptors - Tocris Bioscience
    Rating 4.7 (3) · $45 deliveryNBQX is a selective and competitive AMPA and kainate receptor antagonist (IC50 = 0.15 μM and 4.8 μM, respectively). NBQX blocks the antidepressant effects of 8- ...
  80. [80]
    Talampanel | AMPA Receptors - Tocris Bioscience
    $$45 deliveryTalampanel is a non-competitive AMPA/kainate receptor antagonist that displays 2.3-3-fold more potent activity than GYKI 52466 (Cat. No. 1454).
  81. [81]
  82. [82]
    Positive AMPA and Kainate Receptor Modulators and Their ... - MDPI
    This review presents a comprehensive overview of recent advances in the development of AMPA and kainate receptor PAMs.
  83. [83]
    Interactions of allosteric modulators of AMPA/kainate receptors on ...
    AMPA receptor positive modulator cyclothiazide was more effective in the potentiation of SD evoked by AMPA than by kainate. Slight potentiation of either ...
  84. [84]
    New insights in the development of positive allosteric modulators of ...
    This study explored new AMPAR PAMs belonging to 1,2,4-benzothiadiazine 1,1-dioxides. The introduction of a mono/difluoromethyl side chain at the 2-position was ...
  85. [85]
    GluK2 kainate receptor subunit-selective, potentiating RNA aptamer
    Sep 11, 2025 · Our study demonstrates a potential utility of combining molecular docking with functional assay for a template-based discovery of potent, single ...Missing: potentiators Nature
  86. [86]
    The Antiseizure Drug Perampanel Is a Subunit-Selective Negative ...
    Jul 13, 2022 · Perampanel (PMP) is a third-generation antiseizure drug reported to be a potent and selective noncompetitive negative allosteric modulator of one subfamily of ...
  87. [87]
    Positive AMPA and Kainate Receptor Modulators and Their ...
    This review presents a comprehensive overview of recent advances in the development of AMPA and kainate receptor PAMs. ... mood disorders based on glutamate/GABA ...
  88. [88]
    for the BGG492 Study Group - Sage Journals
    Aug 20, 2013 · Furthermore, the AMPA/kainate receptor antagonist, LY293558, has demonstrated therapeutic benefits in acute migraine (9). BGG492 is an orally ...
  89. [89]
    Therapeutic Potential of Kainate Receptors - Wiley Online Library
    Dec 6, 2010 · However, current Phase II clinical trials using tezampel, an AMPA/receptor antagonist also named NGX424, reduced migraine pain, and effect ...Kars Pharmacology · Kars And Cell Death · Relevance Of Kars And Their...<|separator|>