GRIK2 Antibody, HRP conjugated is a monoclonal antibody chemically linked to HRP, enabling chemiluminescent or colorimetric detection in assays like Western blot (WB) and immunohistochemistry (IHC). It specifically binds to GRIK2 (also termed GluK2 or GLUR6), a kainate-type ionotropic glutamate receptor critical for synaptic transmission and neurophysiological processes .
Immunogen: Synthetic peptide corresponding to amino acids 50–150 of human GRIK2 .
Cross-reactivity: Validated for human, mouse, and rat samples .
Observed molecular weight: ~100–109 kDa, consistent with GRIK2’s predicted size .
Validation data:
| Sample Type | Blocking Buffer | Exposure Time | Band Size | Reference |
|---|---|---|---|---|
| Human hypothalamus | 5% NFDM/TBST | 48 seconds | 109 kDa | |
| Mouse brain | 3% nonfat milk/TBST | 10 seconds | 100 kDa |
Protocol: Heat-mediated antigen retrieval with Tris-EDTA (pH 9.0) or citrate buffer (pH 6.0) .
Positive staining: Observed in human colon carcinoma, kidney, and mouse cerebellum tissues .
GRIK2 is implicated in bipolar disorder (BPD) and cognitive disabilities. Knockout mice exhibit hyperactivity and altered anxiety responses, modulated by lithium treatment .
Dysregulation of GRIK2 is linked to glutamate signaling abnormalities in cancer, including renal and gastric carcinomas .
GRIK2 (Glutamate Receptor, Ionotropic, Kainate 2), also known as GluK2 or GluR6, is a kainate receptor subunit that functions as a glutamate-gated cation channel with diverse roles in the central nervous system. It has gained significant research interest due to its involvement in various neurological conditions and processes. Recent evidence suggests a unique role for GluR6 in controlling abnormalities related to behavioral symptoms of mania, making it a valuable target for understanding certain neuropsychiatric disorders . Additionally, bi-allelic loss of function of the KAR-encoding gene variants are likely pathogenic in certain neurological conditions, highlighting its importance in normal neuronal function .
Based on current literature and commercial offerings, GRIK2 antibodies are available in several configurations:
| Antibody Type | Host Species | Clonality | Common Applications | Example Catalog Numbers |
|---|---|---|---|---|
| Anti-GRIK2/GluK2 | Rabbit | Polyclonal | WB, ELISA | ab28697 |
| Human GluR6/GRIK2 | Mouse | Monoclonal (IgG 2A) | Flow Cytometry, cell-based ELISA | MAB9610 |
| GRIK2 | Rabbit | Monoclonal | WB, IHC | ABIN7267415 |
| GRIK2 (various epitopes) | Rabbit | Polyclonal | WB, IHC, IF, IP, ELISA | Multiple variants |
The selection of an appropriate antibody depends on the specific experimental application, target species, and epitope of interest .
Antibody conjugations, including HRP (horseradish peroxidase), significantly impact application methodologies and detection sensitivity. For GRIK2 research, while many primary antibodies are available unconjugated, HRP conjugation offers direct detection without secondary antibodies in applications like western blotting and ELISA.
The primary advantages of HRP-conjugated GRIK2 antibodies include:
Elimination of secondary antibody incubation steps, reducing protocol time
Minimization of background signal often associated with secondary antibodies
Direct quantification of target proteins
Compatible with multiple detection systems including enhanced chemiluminescence (ECL)
When working with unconjugated primary antibodies, researchers typically use HRP-linked secondary antibodies (anti-rabbit or anti-mouse IgG) as demonstrated in published protocols for GRIK2 detection .
GRIK2 antibodies have demonstrated utility across multiple experimental platforms in neuroscience research:
These applications provide complementary data for understanding GRIK2 expression, localization, and function in normal and pathological conditions .
Rigorous validation of GRIK2 antibodies is essential for reliable experimental outcomes. Methodological approaches include:
Positive and negative control samples:
Peptide competition assays:
Multiple antibody concordance:
Verification with alternative antibodies targeting different epitopes
Comparison of staining/binding patterns across techniques
Isotype controls:
These validation steps should be documented before undertaking extensive experimental work with any GRIK2 antibody .
When using HRP-conjugated GRIK2 antibodies for Western blotting, several methodological adaptations are needed compared to standard two-step detection protocols:
Sample preparation:
Blocking optimization:
Antibody dilution and incubation:
Washing procedure:
More stringent washing may be required (4-6 washes with TBS-T)
Increase washing duration to minimize background
Detection considerations:
These adaptations help maintain sensitivity while reducing background when using directly conjugated antibodies .
Effective immunohistochemical detection of GRIK2 in brain tissue requires specific methodological considerations:
Tissue preparation:
Freshly prepared 4% paraformaldehyde fixation is preferable
Post-fixation time should be optimized to preserve epitope accessibility
For paraffin-embedded sections, antigen retrieval is critical (citrate buffer pH 6.0 or EDTA buffer pH 9.0)
Antibody selection:
Signal amplification:
For HRP-conjugated antibodies, tyramide signal amplification can enhance sensitivity
For unconjugated antibodies, biotin-streptavidin systems or polymer detection methods are effective
Controls and quantification:
Include positive control tissues with known GRIK2 expression
Quantification should utilize standardized imaging parameters and analysis software (e.g., ImageJ)
For co-localization studies, spectral unmixing may be necessary to distinguish signals
Protocol optimization:
Titrate antibody concentration to minimize background
Extended incubation times at 4°C may improve signal-to-noise ratio
Permeabilization conditions should be optimized for access to different cellular compartments
These methodological considerations enable reliable visualization of GRIK2 distribution in neuronal tissues .
Non-specific binding is a common challenge with GRIK2 antibodies that can be addressed through systematic troubleshooting:
Antibody validation:
Blocking optimization:
Antibody incubation conditions:
Reduce antibody concentration (perform titration series)
Add 0.1-0.2% BSA to antibody dilution buffer
For problematic samples, pre-adsorb antibody with tissue powder from the species being tested
Washing optimization:
Increase wash buffer stringency (add 0.2-0.5M NaCl)
Extend washing times and number of washes
Use detergent combinations (Tween-20 plus Triton X-100)
Cross-reactivity assessment:
Perform Western blot to identify potential cross-reactive proteins
Consider alternative antibodies targeting different epitopes
Systematic implementation of these approaches can significantly improve signal specificity .
Quantitative analysis of GRIK2 expression requires appropriate statistical methodologies:
Western blot quantification:
Statistical tests:
Experimental design considerations:
Minimum sample sizes should be determined through power analysis
Technical replicates (n=3 minimum) should be performed
Biological replicates across independent experiments are essential for reliability
Data presentation:
Multiple testing correction:
These statistical approaches ensure rigorous analysis and interpretation of GRIK2 expression data .
Investigation of GRIK2 trafficking and membrane localization requires specialized approaches:
Surface biotinylation assay:
HEK293-T/17 cells transfected with GluK2a cDNA provide a controlled system
Incubate cells with 0.5 mg/mL biotin in cold PBS for 30 min at 4°C with gentle agitation
Quench with 100 mM glycine in cold PBS for 5 min at 4°C
Extract proteins using TEEN-TX buffer (50 mM Tris, 1 mM EDTA, 1 mM EGTA, 150 mM NaCl, 0.1% Triton X-100) with protease inhibitors
Pull down biotinylated proteins and analyze by Western blotting with GRIK2 antibodies
Live-cell imaging approaches:
Combine GRIK2 antibodies recognizing extracellular epitopes with fluorescent secondary antibodies
Monitor trafficking in real-time using confocal microscopy
For pulse-chase experiments, use temperature blocks to synchronize receptor movement
Co-localization studies:
Combine GRIK2 antibodies with markers for specific cellular compartments
Quantify co-localization using Pearson's or Mander's coefficients
Super-resolution microscopy can provide nanoscale resolution of receptor clustering
Receptor internalization assays:
Label surface receptors at 4°C, then allow internalization at 37°C
Strip remaining surface antibodies using acid wash
Quantify internalized receptors by microscopy or flow cytometry
These methodologies enable detailed characterization of GRIK2 trafficking dynamics and membrane expression .
Studying GRIK2 interactions within neural signaling complexes requires sophisticated methodological approaches:
Co-immunoprecipitation strategies:
Extract proteins under non-denaturing conditions to preserve native interactions
Use GRIK2 antibodies for pull-down experiments followed by Western blotting for potential interacting partners
Validate interactions using reciprocal co-immunoprecipitation
Proximity ligation assay (PLA):
Utilize GRIK2 antibodies in combination with antibodies against potential interacting proteins
This technique allows visualization of protein-protein interactions with subcellular resolution
Quantification of PLA signals provides semi-quantitative assessment of interaction strength
BiFC (Bimolecular Fluorescence Complementation):
Genetic approach complementing antibody-based methods
Fusion of split fluorescent proteins to GRIK2 and potential interactors
Reconstitution of fluorescence occurs upon protein-protein interaction
Mass spectrometry approaches:
Following immunoprecipitation with GRIK2 antibodies, analyze pulled-down complexes by LC-MS/MS
Filter results against appropriate controls to identify specific interactors
Validation of novel interactions using orthogonal methods is essential
Advanced imaging techniques:
FRET (Fluorescence Resonance Energy Transfer) using antibody-based fluorophore systems
STED or STORM super-resolution microscopy for nanoscale colocalization analysis
Single-particle tracking to analyze dynamic interactions
These approaches provide complementary data on GRIK2 interactions, enabling comprehensive characterization of its role in signaling complexes .
GRIK2 antibodies can be instrumental in characterizing pathological alterations in neurological disorders through several methodological approaches:
Post-mortem tissue analysis:
Comparative immunohistochemistry and Western blotting across control and disease tissues
Quantification of expression level changes in specific brain regions
Assessment of subcellular distribution alterations in disease states
Animal model characterization:
Analysis of GRIK2 expression in genetic or pharmacological models of neurological disorders
Correlation of expression changes with behavioral phenotypes
Longitudinal studies to track progression of alterations
Patient-derived cell models:
iPSC-derived neurons from patients with GRIK2 mutations
Immunocytochemistry and biochemical analysis of receptor expression and trafficking
Correlation of cellular phenotypes with clinical manifestations
Pharmacological manipulation:
Use of GRIK2 antibodies to monitor receptor responses to therapeutic compounds
Assessment of receptor internalization, phosphorylation, or degradation following drug treatment
Combination with functional assays (calcium imaging, electrophysiology) to correlate structure with function
Genetic validation:
Analysis of GRIK2 expression in genetically defined cases (e.g., clustered mutations in the GRIK2 gene associated with neurodevelopmental disorders)
Correlation of genetic variants with protein expression patterns
Structure-function relationships in receptors containing pathogenic variants
These applications provide critical insights into the role of GRIK2 in neurological disease pathogenesis and potential therapeutic targeting .