The GRIA2/GRIA3 recombinant monoclonal antibody is a highly specific antibody that can target two closely related proteins GRIA2 and GRIA3. It is engineered using advanced biotechnological techniques, such as genetic engineering and antibody engineering. It is produced through the cloning of specific DNA sequences encoding the GRIA2/GRIA3 antibody heavy and light chains into a plasmid vector and subsequent transfection of the recombinant vector into a host cell for expression. The resulting GRIA2/GRIA3 recombinant monoclonal antibody is purified from affinity chromatography from the cell culture supernatant. It has been validated to detect human GRIA2 and GRIA3 in ELISA and IF applications.
The GRIA2/GRIA3 Recombinant Monoclonal Antibody is a highly specific antibody designed to target two closely related proteins, GRIA2 and GRIA3. This antibody is engineered using advanced biotechnological techniques, including genetic engineering and antibody engineering. The production process involves cloning specific DNA sequences encoding the GRIA2/GRIA3 antibody heavy and light chains into a plasmid vector. This recombinant vector is then transfected into a host cell for expression. The resulting GRIA2/GRIA3 Recombinant Monoclonal Antibody is purified using affinity chromatography from the cell culture supernatant. It has been validated for its ability to detect human GRIA2 and GRIA3 in ELISA and immunofluorescence (IF) applications.
GRIA2 and GRIA3 are subunits of AMPA receptors, which belong to the glutamate receptor family. These receptors mediate fast excitatory synaptic transmission at virtually all central synapses, making their functional characteristics critical determinants of brain function . The GRIA2 protein (also known as GluR-2, GluR-B, GluR-K2, GluA2) and GRIA3 protein (also known as GluR-3, GluR-C, GluR-K3, GluA3) are essential components of these receptors . They are significant research targets because mutations in these genes have been associated with various neurological disorders including epilepsy, intellectual disability, autism spectrum disorders, and structural brain abnormalities .
GRIA2/GRIA3 recombinant monoclonal antibodies are engineered using advanced biotechnological techniques. The production process involves cloning specific DNA sequences that encode the antibody's heavy and light chains into a plasmid vector, followed by transfection of this recombinant vector into a host cell (typically HEK293F cells) for expression . After expression, the antibody is purified from the cell culture supernatant using affinity chromatography techniques . This recombinant approach ensures consistency, specificity, and high-quality antibody production compared to traditional hybridoma-based methods.
Based on current validation data, GRIA2/GRIA3 recombinant monoclonal antibodies have been successfully used in enzyme-linked immunosorbent assays (ELISA) and immunofluorescence (IF) applications . For immunofluorescence applications, the recommended dilution range is typically 1:20-1:200 . These antibodies have been specifically validated to detect human GRIA2 and GRIA3 proteins, making them valuable tools for studying these receptors in various research contexts .
RNA editing is a post-transcriptional modification process that alters the nucleotide sequence of RNA molecules. In GRIA genes, important editing sites include the Q/R site and the R/G site . The R/G editing site is found in GRIA2, GRIA3, and GRIA4 subunits . This editing process is developmentally regulated, with editing frequencies increasing during development. For example, the R/G site of GRIA3 shows approximately 15% editing at embryonic day 15 (E15), gradually increasing to 91% at postnatal day 21 (P21) . These editing events can significantly affect receptor function, trafficking, and pharmacological properties, making them important determinants of neurophysiological processes.
Research has identified numerous pathogenic variants in GRIA2 and GRIA3 genes that affect receptor function through multiple mechanisms. These variants can alter the agonist EC50 (concentration producing half-maximal response), response time course, receptor desensitization properties, and/or surface expression levels . For instance, specific variants like p.Glu508Val in GRIA2 (located in the ABD-S1 domain) or p.Met706Leu in GRIA3 (located in the ABD-S2 domain) can disrupt receptor biophysical properties . The functional changes induced by these variants likely contribute to circuit dysfunction, which manifests as clinical phenotypes such as epilepsy, intellectual disability, or autism spectrum disorders. Each variant may have unique effects on receptor function, necessitating detailed functional characterization to understand their pathophysiological mechanisms.
For studying specific GRIA3 variants such as G833R and W637S (associated with Wu Syndrome), a comprehensive in vitro approach is recommended . This approach typically involves:
Cell line selection: Multiple cell types can be used, including HEK293T cells (cultured in DMEM with 10% FBS and 1% penicillin/streptomycin), SH-SY5Y cells (cultured in DMEM with 15% FBS, antibiotics, L-glutamine, and sodium pyruvate), or neural cell lines that provide a relevant cellular context .
Genetic modification: CRISPR/Cas9 or lentiviral-based approaches can be used to introduce the variants of interest.
Verification of expression: Flow cytometry using GRIA3-specific antibodies (such as LSBio Monoclonal Mouse anti-Rat GRIA3/iGLUR3 Antibody) can confirm expression levels .
Functional analysis: Electrophysiological approaches (patch-clamp), calcium imaging, or other functional assays can assess the impact of these variants on receptor function.
This multi-faceted approach provides a robust platform for characterizing the functional consequences of GRIA3 variants.
When using a dual-specificity antibody that recognizes both GRIA2 and GRIA3, differentiating between the effects on these two proteins requires complementary experimental strategies:
Genetic manipulation: Selective knockdown or knockout of either GRIA2 or GRIA3 using siRNA, shRNA, or CRISPR/Cas9 can help determine the contribution of each protein.
Isoform-specific PCR: Design primers that specifically target unique regions of either GRIA2 or GRIA3, as demonstrated in primers used for GRIA3 exon amplification .
Comparison with single-specificity antibodies: Parallel experiments using antibodies specific for only GRIA2 or only GRIA3 can provide validation.
Careful experimental design: Include appropriate controls in immunofluorescence or Western blot experiments to account for potential cross-reactivity.
Mass spectrometry: For definitive protein identification, mass spectrometry-based approaches can distinguish between GRIA2 and GRIA3 based on unique peptide sequences.
RNA editing at the R/G site shows distinct developmental patterns in GRIA2 and GRIA3. In GRIA2, R/G site editing increases from approximately 4% at embryonic day 15 (E15) to 72% at postnatal day 21 (P21) . GRIA3 follows a similar pattern but starts at a higher baseline, with R/G site editing increasing from 15% at E15 to 91% at P21 .
Gene | Editing Site | E15 | P21 | Developmental Change |
---|---|---|---|---|
GRIA2 | R/G | 4% | 72% | +68% |
GRIA3 | R/G | 15% | 91% | +76% |
To accurately measure these editing levels, several techniques can be employed:
Next-generation sequencing (such as 454 Amplicon Sequencing) provides the most accurate quantification of editing frequencies .
Direct PCR sequencing is less sensitive, as editing levels below 20% may not be reliably detected .
Site-specific primers that differentiate between edited and non-edited transcripts can be used for quantitative PCR approaches.
RNA-seq analysis with appropriate bioinformatic pipelines can also detect editing events genome-wide.
The choice of technique depends on the required sensitivity, sample availability, and research question specificity.
For optimal immunofluorescence results with GRIA2/GRIA3 recombinant monoclonal antibodies, follow these methodological guidelines:
Dilution range: Use a dilution between 1:20 and 1:200, with the optimal dilution requiring empirical determination for each application and tissue type .
Fixation protocol: Standard fixation with 4% paraformaldehyde (10-15 minutes) works well for most neural tissues and cell cultures.
Permeabilization: When detecting intracellular epitopes, permeabilize samples with 0.1-0.3% Triton X-100 or use commercial fixation/permeabilization kits (such as BD Cytofix/Cytoperm) .
Blocking: Use 5-10% normal serum (from the species in which the secondary antibody was raised) to minimize non-specific binding.
Antibody incubation: Incubate primary antibody overnight at 4°C for optimal binding and signal-to-noise ratio.
Controls: Always include appropriate negative controls (omitting primary antibody) and positive controls (tissues known to express the target proteins).
Signal detection: Use fluorophore-conjugated secondary antibodies compatible with available imaging systems.
Image acquisition: Capture images using confocal or high-resolution fluorescence microscopy with appropriate filter sets.
Thorough validation of GRIA2/GRIA3 antibody specificity is crucial for experimental rigor and reproducibility. A comprehensive validation strategy includes:
Western blot analysis: Verify that the antibody detects bands of the expected molecular weight (approximately 100 kDa for both GRIA2 and GRIA3).
Peptide competition assays: Pre-incubation of the antibody with the immunizing peptide should eliminate or significantly reduce the specific signal.
Knockout/knockdown controls: Use CRISPR/Cas9-generated GRIA2 or GRIA3 knockout cells or siRNA-mediated knockdown to confirm signal specificity.
Comparison with other validated antibodies: Use multiple antibodies targeting different epitopes of GRIA2 and GRIA3 to confirm staining patterns.
Flow cytometry: As demonstrated in research on GRIA3 variants, flow cytometry can quantitatively assess antibody specificity and protein expression levels .
Immunoprecipitation followed by mass spectrometry: This approach can definitively confirm the identity of the proteins recognized by the antibody.
Testing in multiple cell types with known expression profiles: Compare antibody performance across cell lines with documented GRIA2/GRIA3 expression levels.
For accurate quantification of GRIA2/GRIA3 expression by flow cytometry, the following protocol is recommended:
Cell preparation: Collect approximately 100,000 cells per sample and wash with staining buffer .
Fixation/permeabilization: If detecting intracellular epitopes, use a commercial kit like BD Cytofix/Cytoperm Fixation/Permeabilization Kit .
Antibody staining: Incubate cells with GRIA2/GRIA3 antibody (such as LSBio Monoclonal Mouse anti-Rat GRIA3/iGLUR3 Antibody) for 30 minutes at 4°C .
Washing: Add 1000 μl of staining buffer and centrifuge at 400g for 5 minutes .
Resuspension: Dissolve the pellet in 200 μl of staining buffer .
Analysis: Analyze samples using a flow cytometer (such as Beckman Coulter CytoFlex) to determine both the percentage of positive cells and the Mean Fluorescence Intensity (MFI) .
Controls: Include isotype controls, unstained samples, and single-color controls for accurate gating and compensation.
Data reporting: Report both the percentage of positive cells and MFI, as both measures provide important information about expression levels.
This protocol can be adapted for different cell types by adjusting incubation times and antibody concentrations based on specific cell characteristics.
When faced with contradictory results between immunofluorescence (IF) detection of GRIA2/GRIA3 and functional studies, consider the following analytical approach:
Distinguish between expression and function: Immunofluorescence detects protein presence but does not directly measure functionality. Some variants may affect function without altering expression levels or subcellular localization .
Consider post-translational modifications: RNA editing at sites like R/G can alter receptor function without changing antibody recognition . Verify whether the antibody epitope includes regions subject to editing or other modifications.
Evaluate surface expression versus total protein: Standard IF may detect total cellular protein, while functional studies depend on properly folded, assembled, and cell-surface-expressed receptors. Consider using surface biotinylation or non-permeabilized staining to specifically examine surface expression.
Examine receptor stoichiometry: AMPA receptors are heterotetramers, and altered subunit composition can affect function even when individual subunits are expressed normally.
Assess temporal factors: Developmental regulation of editing and expression may lead to time-dependent discrepancies . Ensure that both methods are examining the same developmental timepoint.
Re-examine antibody specificity: Confirm that the antibody is not cross-reacting with other glutamate receptor subunits or detecting truncated, non-functional protein products.
Studying disease-associated GRIA2/GRIA3 variants presents several technical challenges:
Low variant frequency: Many pathogenic variants are rare, making natural samples difficult to obtain. Solution: Generate recombinant expression systems or use CRISPR/Cas9 to introduce variants into cell lines or primary cultures .
Heterozygosity effects: Many patients are heterozygous for variants, making it difficult to isolate variant effects. Solution: Create cell lines expressing defined ratios of wild-type and variant receptors to mimic heterozygous conditions.
Subunit interactions: GRIA2 and GRIA3 function within receptor complexes, making isolated studies potentially misleading. Solution: Co-express relevant AMPA receptor subunits to form physiologically relevant receptor assemblies.
Isoform complexity: Alternative splicing creates multiple isoforms (e.g., FLIP and FLOP), complicating interpretation. Solution: Use isoform-specific primers and careful experimental design to account for splice variants .
Developmental context: The impact of variants may depend on developmental stage, particularly given the developmental regulation of RNA editing . Solution: Study variants across multiple developmental timepoints or in developmental model systems.
Functional redundancy: Compensation by other glutamate receptor subunits may mask phenotypes. Solution: Consider combinatorial approaches that examine network-level effects rather than just direct receptor properties.
Translation to in vivo phenotypes: In vitro findings may not directly explain patient phenotypes. Solution: Complement cell-based studies with animal models that recapitulate human variants.
Determining the causal relationship between GRIA2/GRIA3 editing abnormalities and disease phenotypes requires a multifaceted approach:
Temporal analysis: Establish whether editing changes precede symptom onset by examining pre-symptomatic samples or developmental timepoints .
Genetic modification: Use CRISPR/Cas9 to modify editing sites to non-editable sequences and observe resulting phenotypes in cellular or animal models.
Correlation analysis: Perform detailed correlation studies between editing levels and phenotype severity across a spectrum of patients.
Mechanistic studies: Elucidate the molecular consequences of altered editing (e.g., changes in calcium permeability, desensitization kinetics) and link these to specific disease manifestations .
Rescue experiments: Attempt to rescue disease phenotypes by restoring normal editing levels through manipulation of editing enzymes (ADAR, ADARB1) or engineered pre-edited mRNA.
Pharmacological modulation: Test whether AMPAR-selective modulators can ameliorate phenotypes despite editing abnormalities, which would suggest editing changes may be nonessential to the disease process .
Population studies: Examine whether editing changes segregate with disease in family studies or large population cohorts.
This comprehensive approach can help establish whether editing abnormalities are primary drivers of disease or secondary consequences of other pathological processes.