GRIK2 Antibody, Biotin conjugated

Shipped with Ice Packs
In Stock

Description

Overview of GRIK2 Antibody, Biotin-Conjugated

GRIK2 antibodies target the kainate subtype of ionotropic glutamate receptors, which mediate excitatory neurotransmission. Biotin conjugation enhances detection sensitivity in assays by leveraging the strong biotin-streptavidin interaction. While the provided search results primarily describe unconjugated GRIK2 antibodies, biotinylation is typically achieved through site-specific conjugation technologies like GlyCLICK (see Section 4) .

Biotin Conjugation Methodology

  • GlyCLICK Technology: Enables site-specific biotin conjugation to IgG antibodies via Fc glycan remodeling, ensuring a degree of labeling (DOL) of 2 .

  • Advantages: Homogeneous conjugates, preserved immunoreactivity, enhanced signal-to-noise ratio in assays .

Recommended Dilutions

ApplicationDilution RangeExample Source
Western Blot1:500–1:6000 Proteintech 13597-1-AP
ELISATitration-dependentAntibodies-Online

Published Findings

  • Western Blot: Detected GRIK2 in human brain tissue (93–120 kDa) and pig cerebellum (103 kDa) .

  • Immunofluorescence: Used to study dopaminergic differentiation in retinal pigment epithelial cells .

Biotin Conjugation Protocol (GlyCLICK)

StepDescription
DeglycosylationRemoves Fc glycans using GlycINATOR .
Azide ActivationIncorporates azide groups via UDP-GalNAz and GalT(Y298L) .
Click ReactionBiotin (sDIBO) conjugated via copper-free strain-promoted cycloaddition .

Storage: Conjugates stable at 4°C for 1 year; avoid freezing .

Comparative Analysis of GRIK2 Antibodies

Catalog No.Host/ClonalityReactivityConjugationApplications
ABIN5611208 Mouse monoclonalHumanUnconjugatedWB, ELISA, IHC
13597-1-AP Rabbit polyclonalHuman, mouse, ratUnconjugatedWB, IF, ELISA
66631-2-Ig Mouse monoclonalHuman, mouse, pigUnconjugatedWB, ELISA

Biotin conjugation is typically performed post-purchase using kits like GlyCLICK Biotin (Genovis) .

Research Implications

Biotin-conjugated GRIK2 antibodies enable high-sensitivity detection in:

  • Neurodegenerative Studies: Tracking receptor expression in brain tissues .

  • Drug Development: Validating targets in neurological disorders.

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship the products within 1-3 business days after receiving your order. Delivery time may vary depending on the purchase method or location. Please consult your local distributor for specific delivery times.
Synonyms
bA487F5.1 antibody; EAA4 antibody; Excitatory amino acid receptor 4 antibody; GLR 6 antibody; GLR6 antibody; GluK2 antibody; GLUK6 antibody; GLUR 6 antibody; GluR-6 antibody; GluR6 antibody; Glutamate receptor 6 antibody; Glutamate receptor antibody; glutamate receptor form A antibody; glutamate receptor form B antibody; glutamate receptor form C antibody; glutamate receptor form D antibody; glutamate receptor form E antibody; Glutamate receptor ionotropic kainate 2 antibody; GRIK 2 antibody; GRIK2 antibody; GRIK2 protein antibody; GRIK2_HUMAN antibody; ionotropic kainate 2 antibody; MRT6 antibody
Target Names
Uniprot No.

Target Background

Function
The GRIK2 protein is an ionotropic glutamate receptor. L-glutamate serves as an excitatory neurotransmitter at many synapses within the central nervous system. Binding of the excitatory neurotransmitter L-glutamate induces a conformational change, leading to the opening of the cation channel. This conversion transforms the chemical signal into an electrical impulse. The receptor then rapidly desensitizes and enters a transient inactive state, characterized by the presence of bound agonist. GRIK2 modulates the cell surface expression of NETO2, independent of its ionotropic glutamate receptor activity. Additionally, it acts as a thermoreceptor, conferring sensitivity to cold temperatures. GRIK2 functions in dorsal root ganglion neurons.
Gene References Into Functions
  1. Research suggests that PKC SUMOylation plays a crucial role in regulating the 14-3-3 and GluK2a protein complex and may contribute to regulating the decay kinetics of kainate receptor-excitatory postsynaptic currents. PMID: 28837400
  2. These findings indicate that GRIK2 plays a role in the maintenance of urothelial cancer stem cells and that GRIK2 and ALDH1 can serve as prognostic markers for urinary tract carcinomas. PMID: 28418868
  3. TTBK2 down-regulates GluK2 activity by decreasing the receptor protein abundance in the cell membrane via RAB5-dependent endocytosis. PMID: 27607061
  4. In the Han population of Central China, polymorphisms of SNP rs9390754 within the GRIK2 gene may be associated with epilepsy susceptibility. PMID: 27324535
  5. The study demonstrates that co-assembly of recombinant kainate receptors (GluK1 and GluK2) with the Neto1 and Neto2 auxiliary subunits alters their onset and recovery from desensitization in a subunit-dependent manner. PMID: 26277340
  6. This study found that GRIK2 (glutamate receptor, ionotropic kainate 2) showed the most significant correlations with gene expression. PMID: 24662927
  7. This study indicated an association between Gluk2 and obsessive-compulsive disorder. PMID: 24821223
  8. Parkin interacts with the kainate receptor GluK2 subunit and regulates KAR function. PMID: 25316086
  9. High-risk genetic markers of paranoid schizophrenia were identified as GRIK2*ATG and GRIK2*TGG in Tatars. PMID: 25842862
  10. The present study reveals an additional mechanism for the regulation of GluK2-containing kainate receptors by Src family kinases, which may be of pathological significance in ischemic stroke. PMID: 25201974
  11. No association was found between rs1556995 in GRIK2 and clozapine-induced obsessive-compulsive (OC) symptoms, suggesting that GRIK2 may not play a role in the development of OC symptoms in schizophrenia patients. PMID: 23660601
  12. Crosslinking the ligand-binding domain dimer interface locks kainate receptors out of the main open state. PMID: 23713029
  13. 14-3-3 proteins serve as an important regulator of GluK2a-containing KARs and may contribute to the slow decay kinetics of native KAR-EPSCs. PMID: 23861400
  14. Kainate receptor GluK2a post-translational modifications differentially regulate association with 4.1N to control activity-dependent receptor endocytosis. PMID: 23400781
  15. Eight chromosome 6 SNPs, exhibiting the most significant differences, were identified: rs10499298, rs10499299, rs17827966, rs1224329, rs1150790, rs713050, rs2518344, and rs487083; all were associated with the GRIK2 gene. PMID: 23037145
  16. The ACAG haplotype in the 13th haplotype block of the GRIK2 gene was associated with somatic anxiety. PMID: 22429480
  17. Comprehensive analytical methods applied to a much larger sample than in previous studies do not support a role for GRIK2 as a genetic modifier of the age at onset of clinical symptoms in Huntington's disease. PMID: 22771793
  18. Convergent functional genomics allowed the identification of novel candidate genes, GRIK2 and NPAS2, involved in glutamatergic neurotransmission and the circadian rhythm, respectively, that are potentially associated with CFS. PMID: 21912186
  19. Genotyping and linkage analysis excluded linkage of the GRIK2 gene and TUSC3 gene with mental retardation. PMID: 21557188
  20. This study supports previously reported findings of an association between proximal GRIK2 single nucleotide polymorphism and obsessive-compulsive disorder in a comprehensive evaluation of the gene. PMID: 20370803
  21. Human GluK2 is a slowly activating channel but more sensitive to glutamate, compared to the rat ortholog. The M867I mutation does not affect the rate of the equilibrium constants of channel opening but does slow down the channel desensitization rate. PMID: 20863077
  22. Spliced variants of ionotropic glutamate receptor GluR6 are associated with astrocytoma. PMID: 20230879
  23. Phosphorylation of PKC sites on GluR6 regulates surface expression of GluR6 at distinct intracellular trafficking pathways. PMID: 19920140
  24. Linkage and association of the glutamate receptor 6 gene with autism. PMID: 11920157
  25. Specific alleles in the GluR6 and CA150 locus were only observed in HD patients. PMID: 12821179
  26. Describes the time course of the open-channel form of the receptor as a function of glutamate concentration. PMID: 14567698
  27. Using three single nucleotide polymorphisms in GRIK2 and one in GRIK3, no evidence was found for an association with obsessive-compulsive disorder in case-control or family-based analyses. PMID: 15094479
  28. This study examined the maternal transmission disequilibrium of the glutamate receptor GRIK2 in schizophrenia. PMID: 15305151
  29. The modifier effect is actually due to the TAA repeat itself, possibly through a functional consequence on the GRIK2 mRNA. PMID: 16959037
  30. Our data on altered functional properties of GluR6(M836I) provide a functional basis for the postulated linkage of GluR6 to autism. PMID: 17167233
  31. Phosphorylation of the C-terminal tail of GluR6 by PKA leads to potentiation of the whole-cell response. PMID: 17379418
  32. These results suggest a potential association between GRIK2 and autism in the Korean population. PMID: 17428563
  33. Mutations of GluR6 are unlikely to be associated with autism in the Indian population. PMID: 17712621
  34. A complex mutation in the ionotropic glutamate receptor 6 gene (GRIK2, also called "GLUR6") that cosegregates with moderate-to-severe nonsyndromic autosomal recessive mental retardation is reported in a large, consanguineous Iranian family. PMID: 17847003
  35. Overall, our data indicate that hGluR6c might have unique properties in non-nervous cells and in the first stages of CNS development. PMID: 18289788
  36. The apo state of GluR6 undergoes a cleft closure of 29-30 degrees upon binding full agonists, one of the largest observed in the glutamate receptor family. PMID: 18658129
  37. Mutations to the GLUR6 binding pocket that selectively affect domoate binding are reported. PMID: 18664604
  38. Nonconserved residues in GluR6 define the size of the agonist-binding pocket, exerting a steric influence on the bound agonist and the extent of binding-domain closure. PMID: 18690046
  39. GluR6 C-terminal domain KRIP6 regulates kainate receptors by inhibiting PICK1 modulation via competition or a mutual blocking effect. PMID: 18692513
  40. In silico ligand-docking predicted that most partial agonists select for the closed and not, as expected, the open or intermediate conformations of the GluK2 agonist binding domain. PMID: 19225180
  41. Results indicate that ions can contribute substantial free energy to active state stabilization in GluR6 and provide quantitative measurements of the energetic consequences of allosteric ion binding to a ligand-gated ion channel. PMID: 19617541

Show More

Hide All

Database Links

HGNC: 4580

OMIM: 138244

KEGG: hsa:2898

STRING: 9606.ENSP00000397026

UniGene: Hs.98262

Involvement In Disease
Mental retardation, autosomal recessive 6 (MRT6)
Protein Families
Glutamate-gated ion channel (TC 1.A.10.1) family, GRIK2 subfamily
Subcellular Location
Cell membrane; Multi-pass membrane protein. Cell junction, synapse, postsynaptic cell membrane; Multi-pass membrane protein.
Tissue Specificity
Expression is higher in cerebellum than in cerebral cortex.

Q&A

What is GRIK2 and why is it important in neuroscience research?

GRIK2 (also known as GluK2, GLR6, or GLUR6) is an ionotropic glutamate receptor of the kainate family with a calculated molecular weight of approximately 103 kDa . It plays a crucial role in excitatory neurotransmission in the central nervous system, particularly in the hippocampus. Research has demonstrated its involvement in several neurological processes and pathologies, including neurodevelopmental disorders associated with specific mutations such as the Ala657Thr mutation in the GluK2 receptor subunit . GRIK2 is abundantly expressed in the dentate gyrus and CA3 regions of the hippocampus, making it a significant target for studies of synaptic plasticity, neuronal excitability, and glutamatergic signaling pathways .

What are the advantages of using a biotin-conjugated GRIK2 antibody over unconjugated versions?

Biotin-conjugated antibodies offer several experimental advantages compared to their unconjugated counterparts. The high-affinity interaction between biotin and streptavidin (or avidin) provides enhanced signal amplification through multiple binding sites, significantly improving detection sensitivity in applications like immunohistochemistry, flow cytometry, and ELISA. This conjugation eliminates the need for secondary antibody incubation steps, reducing background signal and simplifying experimental workflows . Additionally, the biotin-streptavidin system allows for flexible detection through various reporter molecules (fluorophores, enzymes) without requiring different secondary antibodies for each application, making the biotin-conjugated GRIK2 antibody a versatile tool for multicolor immunostaining protocols in complex neural tissue sections .

What species reactivity can be expected from GRIK2 antibodies?

Based on available data, GRIK2 antibodies demonstrate varying cross-reactivity patterns depending on their specific clone and production method. Many commercially available GRIK2 antibodies show reactivity with human, mouse, and rat samples . More specifically, the mouse monoclonal GRIK2 antibody from Proteintech (66631-2-Ig) has confirmed reactivity with human, mouse, rat, and pig samples in Western blot applications . Some rabbit polyclonal antibodies demonstrate even broader cross-reactivity, with some variants reacting with samples from human, mouse, rat, dog, cow, guinea pig, horse, rabbit, and even zebrafish tissues . When selecting a GRIK2 antibody for your research, it is essential to verify the specific cross-reactivity pattern for your species of interest, particularly if working with less common model organisms .

What are the optimal application methods for biotin-conjugated GRIK2 antibodies?

Biotin-conjugated GRIK2 antibodies can be employed in multiple experimental applications with specific optimization requirements for each technique. For Western blotting, a dilution range of 1:1000 to 1:6000 is typically recommended, with optimal results often achieved at 1:2000 for detecting the 103-115 kDa GRIK2 protein band . For immunohistochemistry and immunofluorescence, where tissue penetration is crucial, lower dilutions (1:100 to 1:500) may be necessary, along with appropriate antigen retrieval methods specific to fixed neural tissues . In flow cytometry applications, titration experiments starting at 1:50 to 1:200 are advised to determine optimal signal-to-noise ratios. For all applications, it's recommended to include positive controls (cerebellum tissue is particularly suitable) and negative controls (primary antibody omission) to validate staining specificity .

How should I optimize antigen retrieval for GRIK2 immunodetection in fixed neural tissues?

Effective antigen retrieval is critical for successful GRIK2 immunodetection in fixed tissues due to the complex structure of this transmembrane receptor. Heat-induced epitope retrieval (HIER) using citrate buffer (pH 6.0) has proven effective for many GRIK2 epitopes, particularly when performed at 95-98°C for 15-20 minutes . For formalin-fixed paraffin-embedded (FFPE) tissues, extending this time to 25-30 minutes may be necessary. Alternative methods include using Tris-EDTA buffer (pH 9.0) for certain epitopes, especially when working with phosphorylation-specific antibodies. For fresh-frozen tissue sections, milder retrieval methods such as brief protease treatment (0.05% trypsin, 5 minutes at 37°C) may suffice. Following antigen retrieval, a blocking step with an avidin/biotin blocking kit is essential to minimize endogenous biotin interference, particularly in tissues with high endogenous biotin levels such as brain, kidney, and liver .

What controls should be implemented when using GRIK2 antibodies in experimental protocols?

Rigorous control implementation is essential for validating GRIK2 antibody specificity and experimental reliability. Primary controls should include positive tissue controls such as cerebellum tissue, which shows high GRIK2 expression, particularly in granule cells . For negative controls, use tissues from GRIK2 knockout models or siRNA-treated samples where available. If these are not accessible, primary antibody omission controls and isotype controls (using matched IgG at the same concentration) should be implemented . For biotin-conjugated antibodies specifically, endogenous biotin blocking is critical, particularly in tissues with high biotin content like brain samples. Additionally, peptide competition assays, where the antibody is pre-incubated with the immunizing peptide, can confirm binding specificity. When analyzing developmental or disease-specific changes in GRIK2 expression, appropriate age-matched or condition-matched controls are essential for accurate interpretations of experimental findings .

How can I address high background issues when using biotin-conjugated GRIK2 antibodies?

High background is a common challenge when working with biotin-conjugated antibodies in neural tissues due to endogenous biotin and non-specific binding. To address this issue, implement a comprehensive blocking protocol starting with an avidin/biotin blocking kit before applying the primary antibody . Optimize protein blocking by using a combination of 5% normal serum (matching the species of your secondary detection reagent) with 1-3% BSA in PBS or TBS. If background persists, add 0.1-0.3% Triton X-100 to improve antibody penetration while reducing non-specific membrane interactions. Increasing washing steps (at least 3-5 washes of 5-10 minutes each) with PBS-T (0.05% Tween-20) between reagent applications can significantly reduce background. For particularly challenging samples, consider reducing the concentration of the streptavidin detection reagent or implementing a centrifugation step (14,000g for 10 minutes at 4°C) to remove potential aggregates from the antibody solution before application .

What are the most common sources of false negative results when working with GRIK2 antibodies?

False negative results with GRIK2 antibodies can stem from several methodological factors. Insufficient antigen retrieval is a primary concern, particularly with formalin-fixed tissues where GRIK2 epitopes may be masked by crosslinking . If this is suspected, extend heat-induced epitope retrieval time or try alternative buffers such as Tris-EDTA (pH 9.0) instead of citrate buffer. Antibody degradation can also cause false negatives; ensure proper storage at -20°C with glycerol and avoid repeated freeze-thaw cycles (aliquot upon receipt) . Importantly, the specific epitope recognized by your GRIK2 antibody might be absent in certain splice variants or post-translationally modified forms of the protein. The GRIK2 gene encodes multiple splice variants that differ in their C-terminal domains, so antibodies targeting these regions may not detect all isoforms . Additionally, certain experimental conditions such as harsh fixation protocols, excessive detergent concentrations, or protease treatment can destroy GRIK2 epitopes. If encountering persistent negative results despite positive controls working, consider testing alternative GRIK2 antibodies that target different epitopes or employing more sensitive detection methods such as tyramide signal amplification .

How can I verify the specificity of my GRIK2 antibody results?

Verifying antibody specificity is critical for confident interpretation of GRIK2 immunolabeling results. A multi-method validation approach is recommended, beginning with Western blot analysis to confirm detection of the expected 103-115 kDa band pattern characteristic of GRIK2 . For definitive validation, perform parallel experiments using tissues from GRIK2 knockout models or cells treated with GRIK2-specific siRNA to confirm signal elimination. When knockout controls aren't available, peptide competition assays can be informative; pre-incubating the antibody with excess immunizing peptide should substantially reduce or eliminate specific staining . For immunohistochemical applications, compare staining patterns with published GRIK2 mRNA expression data or alternative antibodies targeting different GRIK2 epitopes. Additionally, dual-labeling experiments with established neuronal subtype markers can help confirm expected GRIK2 distribution patterns, particularly in hippocampal regions where GRIK2 expression is well-characterized . For biotin-conjugated antibodies specifically, include parallel experiments using the unconjugated version of the same antibody clone when available to ensure conjugation hasn't affected specificity .

How can I optimize GRIK2 antibodies for studying receptor trafficking and subcellular localization?

Investigating GRIK2 receptor trafficking requires specialized immunofluorescence approaches to visualize subcellular localization with high resolution. For optimal results, use thin tissue sections (5-10 μm) or cultured neurons with minimal background interference. Antibodies recognizing extracellular epitopes of GRIK2 are particularly valuable for distinguishing surface-expressed receptors from internal pools through non-permeabilized immunolabeling protocols . For tracking receptor internalization, combine surface labeling (performed at 4°C to prevent endocytosis) with subsequent temperature shifts to 37°C for defined time periods. Confocal microscopy with colocalization analysis using markers for specific subcellular compartments (PSD-95 for postsynaptic densities, Rab5 for early endosomes, etc.) provides quantitative measures of GRIK2 trafficking . For super-resolution approaches like STORM or PALM, directly conjugated antibodies minimize localization error from secondary antibody displacement. When investigating activity-dependent trafficking, pharmacological manipulations with glutamate receptor agonists/antagonists (e.g., kainate, CNQX) combined with time-course immunocytochemistry can reveal dynamic changes in GRIK2 surface expression .

What are the considerations for using GRIK2 antibodies in co-immunoprecipitation studies of receptor complexes?

Co-immunoprecipitation (co-IP) of GRIK2-containing complexes requires careful optimization to preserve native protein interactions while achieving specific pull-down. Begin with gentle lysis conditions using buffers containing 1% non-denaturing detergents like NP-40 or Triton X-100, supplemented with protease and phosphatase inhibitors to maintain protein integrity . Pre-clearing lysates with protein A/G beads before antibody addition significantly reduces non-specific binding. For biotin-conjugated GRIK2 antibodies, streptavidin-coated magnetic beads provide efficient capture with minimal background compared to traditional agarose beads. Critical control experiments should include IgG-matched controls and, where possible, immunoprecipitation from GRIK2-deficient tissues . When investigating GRIK2 interacting partners, consider the transient nature of some interactions; crosslinking with membrane-permeable crosslinkers (e.g., DSP at 1-2 mM) prior to lysis can stabilize weaker interactions. For detecting post-translational modifications of GRIK2, such as phosphorylation events that regulate channel function, phosphatase inhibitor cocktails must be included in all buffers. Elution conditions should be optimized based on downstream applications - milder elution with competing peptides maintains complex integrity for functional studies, while denaturing elution provides higher yields for mass spectrometry analysis of complex composition .

How should I interpret discrepancies between GRIK2 protein and mRNA expression levels?

Discrepancies between GRIK2 protein levels (detected by antibodies) and mRNA expression are commonly observed in neuroscience research and require careful interpretation. These disparities often reflect post-transcriptional regulatory mechanisms that influence GRIK2 expression. MicroRNAs targeting GRIK2 mRNA can significantly reduce protein translation without affecting transcript detection; for example, miR-92a has been shown to regulate GRIK2 expression in neurons . Additionally, GRIK2 undergoes extensive RNA editing, particularly at the Q/R site, which can influence both protein stability and antibody recognition depending on the epitope targeted . Protein turnover rates for GRIK2 receptors also differ from mRNA degradation kinetics, with receptor half-life influenced by activity-dependent endocytosis and degradation. To properly interpret such discrepancies, integrate multiple methodological approaches, including quantitative PCR for transcript levels, Western blotting for total protein, and surface biotinylation assays for membrane-expressed receptors. When analyzing regional differences in expression, consider cell-type specificity and subcellular compartmentalization, as GRIK2 proteins may be trafficked to dendrites far from the cell body where mRNA is typically detected .

What statistical approaches are recommended for analyzing GRIK2 immunolabeling intensity across experimental groups?

Quantitative analysis of GRIK2 immunolabeling requires rigorous statistical approaches tailored to the experimental design and data distribution. For Western blot densitometry comparing GRIK2 expression across groups, normalized data should be analyzed using parametric tests (t-test or ANOVA) if normally distributed, or non-parametric alternatives (Mann-Whitney or Kruskal-Wallis) if assumptions of normality are violated . When analyzing immunohistochemical or immunofluorescence intensity data, hierarchical statistical approaches are recommended to account for nested variables (multiple measurements per section, multiple sections per animal). Linear mixed models or nested ANOVA designs can appropriately handle this data structure while controlling for random effects. For colocalization analyses of GRIK2 with other neuronal markers, Pearson's or Mander's coefficients provide quantitative measures, though threshold selection critically influences results and should be consistently applied across experimental groups. Power analysis prior to experimentation is essential, particularly when studying subtle changes in receptor expression; detecting a 20% difference in GRIK2 expression typically requires 6-8 animals per group with alpha=0.05 and power=0.8. For all statistical approaches, appropriate corrections for multiple comparisons (e.g., Bonferroni, Tukey, or false discovery rate methods) should be implemented when analyzing GRIK2 expression across multiple brain regions or time points .

How can I differentiate between specific GRIK2 isoforms in my experimental results?

Differentiating between GRIK2 isoforms requires strategic antibody selection and complementary molecular approaches. GRIK2 undergoes extensive alternative splicing, particularly at the C-terminus, generating isoforms with distinct functional properties and molecular weights . To distinguish these variants, first select antibodies targeting isoform-specific epitopes; antibodies recognizing the N-terminal domain (amino acids 30-300) will detect most isoforms, while those targeting the C-terminal region can be isoform-specific . In Western blot applications, careful analysis of molecular weight patterns can help identify specific variants - the primary GRIK2 isoform appears at 103-115 kDa, while splice variants may show subtle size differences . For definitive isoform identification, combine immunoprecipitation with mass spectrometry analysis or isoform-specific RT-PCR. When analyzing RNA editing of GRIK2 (particularly Q/R and I/V sites), specialized approaches are required, as standard antibodies cannot distinguish edited forms. For this purpose, restriction fragment length polymorphism (RFLP) analysis of RT-PCR products provides a quantitative measure of editing efficiency. Additionally, when investigating region-specific expression of GRIK2 isoforms, laser capture microdissection combined with RT-PCR and immunohistochemistry can reveal cell-type specific expression patterns that may be obscured in whole-tissue analyses .

How can GRIK2 antibodies be integrated with new imaging technologies for in vivo receptor dynamics?

The integration of GRIK2 antibodies with emerging imaging technologies offers exciting opportunities for studying receptor dynamics in living systems. For in vivo applications, consider using fluorescently tagged nanobodies derived from GRIK2 antibodies, which penetrate tissue more effectively due to their smaller size (~15 kDa compared to ~150 kDa for conventional antibodies) . These can be introduced through viral vectors for long-term expression or directly injected for acute imaging. For longitudinal studies in transgenic models, knock-in approaches introducing epitope tags (HA, FLAG) into the GRIK2 gene allow for specific antibody recognition without affecting receptor function. Advanced two-photon microscopy combined with cranial window techniques permits repeated imaging of GRIK2 dynamics in the same animals over time, particularly valuable for developmental studies or disease progression models . Emerging expansion microscopy protocols can be optimized for GRIK2 detection by using biotin-conjugated antibodies with subsequent streptavidin-fluorophore labeling, enabling super-resolution imaging with conventional microscopes. For functional correlation, consider combining immunolabeling with activity sensors through spectral unmixing approaches - for example, using GCaMP for calcium imaging simultaneously with red-shifted fluorophores for GRIK2 detection .

What methods are being developed to study GRIK2 post-translational modifications using modified antibodies?

Emerging methodologies for studying GRIK2 post-translational modifications (PTMs) focus on developing modification-specific antibodies and integrating them with advanced proteomics. Phosphorylation-specific GRIK2 antibodies targeting key regulatory sites (particularly serine 868 and serine 892) are being developed to monitor activity-dependent receptor regulation . These phospho-specific antibodies can be integrated with proximity ligation assays (PLA) to visualize specific phosphorylation events with subcellular resolution in fixed tissues. For comprehensive PTM mapping, immunoprecipitation with pan-GRIK2 antibodies followed by mass spectrometry analysis allows identification of multiple modifications simultaneously, including phosphorylation, ubiquitination, SUMOylation, and glycosylation patterns that regulate receptor trafficking and function . Novel biotin-conjugated antibodies with cleavable linkers are being developed to facilitate sequential elution strategies, allowing efficient recovery of GRIK2 complexes for downstream PTM analysis. Time-resolved approaches combining stimulation protocols (e.g., LTP induction) with rapid fixation and phospho-specific immunolabeling can reveal the temporal dynamics of GRIK2 modifications in response to neuronal activity. Additionally, CRISPR-based strategies to introduce specific mutations at PTM sites, combined with antibody detection of resulting functional changes, provide powerful tools for dissecting the roles of individual modifications in GRIK2 function .

How will antibody-based approaches contribute to understanding GRIK2 involvement in neurodevelopmental disorders?

Antibody-based approaches are poised to make significant contributions to understanding GRIK2's role in neurodevelopmental disorders, particularly in light of recent findings linking GRIK2 mutations to these conditions. The A657T mutation in GRIK2, which has been associated with neurodevelopmental disorders, provides a specific target for investigating pathological mechanisms . Comparative immunohistochemistry between wild-type and A657T knock-in mouse models can reveal alterations in receptor localization, particularly in hippocampal circuits where abnormal KAR signaling has been observed . Custom antibodies specifically recognizing the mutant form of GRIK2 would enable direct visualization of the mutant protein's trafficking and accumulation patterns. For human studies, immunohistochemical analysis of post-mortem brain tissue from individuals with GRIK2 mutations could reveal altered expression patterns compared to neurotypical controls . Integrating GRIK2 antibody labeling with electrophysiological recordings in brain slice preparations from disease models provides crucial functional correlates to expression changes. Additionally, biotin-conjugated GRIK2 antibodies can be applied in proteomic studies to identify disrupted protein interactions in the context of disease-associated mutations. As precision medicine approaches evolve, antibody-based diagnostics might enable stratification of neurodevelopmental disorder subtypes based on GRIK2 expression patterns, potentially guiding personalized therapeutic strategies targeting specific glutamatergic signaling disruptions .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.