GRM2 antibodies are used in:
Key findings include:
Paraneoplastic cerebellar ataxia: GRM2 antibodies were identified in patients with subacute ataxia and tumors (e.g., alveolar rhabdomyosarcoma) .
Pathogenic mechanism: Unlike antibodies causing receptor internalization (e.g., anti-NMDAR), GRM2 antibodies may block receptor function without reducing surface expression .
| Case | Age/Sex | Clinical Features | Tumor Association | Immunotherapy Response |
|---|---|---|---|---|
| 1 | 78/Female | Progressive cerebellar ataxia | Small-cell tumor | Poor |
| 2 | 3/Female | Acute cerebellitis, nystagmus | Alveolar rhabdomyosarcoma | Good (steroid-responsive) |
| 3 | 56/Female | Chronic ataxia, brainstem atrophy | None identified | Poor |
Outcomes: Early immunotherapy improved symptoms in pediatric cases but not in adults, suggesting irreversible neuronal damage in chronic stages .
| Product Code | Host | Target Region | Applications | Reactivity |
|---|---|---|---|---|
| ER2/3 | Chicken | Shared mGluR2/mGluR3 | IHC, ICC | Human, Mouse, Rat |
| ABIN1048928 | Rabbit | N-terminal (AA 109–121) | IHC, IHC (paraffin) | Human, Dog, Bat |
Role in synaptic modulation: GRM2 antibodies may disrupt glutamate/GABA balance, leading to cerebellar dysfunction .
Tumor links: mGluR2 is expressed in neoplasms like rhabdomyosarcoma, suggesting a paraneoplastic antigenic role .
GRM2 (also known as GPRC1B, MGLUR2, mGlu2) is a metabotropic glutamate receptor with 872 amino acid residues and a mass of approximately 95.6 kDa. It belongs to the G-protein coupled receptor 3 family and is primarily expressed in the brain cortex, where it participates in chemical synaptic transmission . GRM2 has gained significance in research due to its involvement in multiple neurological functions, spatial memory processes, and potential roles in psychiatric disorders . Additionally, recent research has revealed unexpected functions for GRM2, including its role as an endocytic receptor for influenza virus entry . The development of specific antibodies against GRM2 has been crucial for characterizing its expression patterns, interacting partners, and functional roles across various tissues and experimental models.
GRM2 antibodies typically target various regions including the N-terminal extracellular domain, C-terminal region, and internal regions. Commercial antibodies most frequently target the N-terminal domain (AA 414-558, AA 109-121) or more specific regions such as AA 801-872 . Epitope selection significantly impacts antibody utility:
N-terminal targeting antibodies (extracellular domain): Ideal for cell-surface detection, receptor blocking studies, and immunoprecipitation of intact protein. These antibodies can detect native conformations and are particularly useful for functional studies involving receptor-ligand interactions .
C-terminal targeting antibodies: Better suited for Western blot applications after protein denaturation, especially when studying truncated variants or post-translational modifications of the intracellular domain .
Internal region antibodies: Useful for detecting proteolytic fragments or studying domain-specific functions .
When selecting a GRM2 antibody, researchers should consider which region best suits their experimental questions, as epitope choice directly affects detection sensitivity and experimental outcomes.
Distinguishing between GRM2 and GRM3 presents a significant challenge due to their structural similarity. According to specificity analyses, some commercial GRM2 antibodies show up to 50% homology with GRM3 . To ensure specific detection:
BLAST analysis validation: Before selecting an antibody, review the manufacturer's BLAST analysis of the peptide immunogen. Antibodies targeting unique regions show minimal homology with GRM3 .
Knockout validation: Use tissues/cells from GRM2 knockout models as negative controls to confirm antibody specificity. In a study examining GRM2 function in adult-born dentate gyrus cells, researchers validated antibody specificity using knockout controls .
Cross-reactivity testing: If possible, test reactivity against recombinant GRM2 and GRM3 proteins in parallel experiments.
Western blot analysis: While both proteins have similar molecular weights, slight differences in migration patterns can help distinguish them. GRM2 typically appears at approximately 95.6 kDa .
Combination with mRNA analysis: Complement antibody detection with qPCR or RNAscope to verify that protein detection correlates with mRNA expression patterns.
When absolute specificity is required, researchers should consider using antibodies targeting the most divergent regions between GRM2 and GRM3, or employing genetic approaches like shRNA knockdown to confirm detection specificity .
Achieving optimal Western blot results with GRM2 antibodies requires careful consideration of several parameters:
Sample Preparation:
Brain tissue samples should be homogenized in RIPA buffer containing EDTA-free protease inhibitors and phosphatase inhibitors on ice .
Centrifugation at 12,000 rpm at 4°C for 15 minutes is recommended to separate cellular debris .
For cultured cells, washing with cold PBS followed by lysis in RIPA buffer for 10-20 minutes provides optimal protein extraction .
Running Conditions:
Use 7.5-10% SDS-PAGE gels to achieve proper separation of the 95.6 kDa GRM2 protein.
A longer running time may help distinguish GRM2 from other similar-sized proteins.
Antibody Dilutions:
Primary antibody concentrations vary by manufacturer but typically range from 1:500 (e.g., Sigma-Aldrich SAB4501319) to 1:1000 (e.g., Boster Bio A06123-3) .
Secondary antibody dilutions generally range from 1:3000 to 1:5000 depending on detection method .
Membrane Incubation:
Overnight incubation at 4°C with primary antibody provides optimal binding .
One-hour room temperature incubation with secondary antibody is typically sufficient .
Detection Controls:
For stringent validation, include a GRM2 knockout sample or tissue known to lack GRM2 expression.
Following these conditions helps ensure specific detection of GRM2 while minimizing background and cross-reactivity issues that can complicate interpretation of results.
Optimizing immunohistochemistry (IHC) for GRM2 detection requires attention to several critical parameters:
Fixation and Embedding:
Formalin fixation and paraffin embedding represent the standard approach for GRM2 IHC .
Section thickness should be approximately 4 μm for optimal antibody penetration and signal resolution .
Antigen Retrieval:
Heat-induced epitope retrieval is typically necessary due to the membrane-bound nature of GRM2.
Citrate buffer (pH 6.0) or EDTA buffer (pH 9.0) are commonly used, with the optimal choice depending on the specific antibody epitope.
Antibody Dilutions:
For commercial antibodies, recommended dilutions range from 1:50-1:200 to 32 μg/mL depending on the manufacturer and specific antibody sensitivity.
Optimization through dilution series is advised for each new tissue type or fixation method.
Background Reduction:
Permeabilize with 0.2% Triton X-100 and block with 5% normal donkey serum to minimize non-specific binding .
For brain tissue with high endogenous peroxidase activity, hydrogen peroxide treatment is recommended before antibody incubation.
Visualization Methods:
For brightfield microscopy, horseradish peroxidase-conjugated secondary antibodies with DAB substrate provide reliable detection.
For fluorescence, secondary antibodies should be carefully selected to avoid spectral overlap with other markers in co-labeling experiments.
Validation Controls:
Include positive tissue controls (brain cortex sections) where GRM2 is known to be highly expressed .
Negative controls should include primary antibody omission and, ideally, GRM2 knockout tissue.
A methodical approach to optimization will yield reliable and reproducible GRM2 detection in histological preparations.
When using GRM2 antibodies for functional blocking experiments, researchers should consider several critical factors:
Antibody Selection Criteria:
Choose antibodies specifically targeting the extracellular N-terminal domain of GRM2, as these can access the receptor in its native conformation on the cell surface .
Monoclonal antibodies often provide more consistent blocking results compared to polyclonal antibodies due to their defined epitope specificity .
Validation of Blocking Efficiency:
Before conducting the main experiment, verify the antibody's blocking capacity using positive controls.
In studies examining GRM2's role as an endocytic receptor for influenza virus, researchers validated blocking efficiency using a dose-dependent neutralization assay .
Experimental Design Considerations:
Include appropriate isotype controls to distinguish specific blocking effects from non-specific antibody binding.
Determine effective antibody concentrations through dose-response experiments. Published studies have demonstrated that monoclonal antibodies against the ectodomain of GRM2 (mGluR2-ma) can inhibit H1N1 virus infection in a dose-dependent manner .
Consider potential complement activation or Fc receptor engagement, which might confound interpretation of blocking experiments.
Alternative Approaches for Validation:
Compare antibody blocking results with soluble receptor competition assays. For example, purified GRM2 ectodomain with a GST tag (mGluR2-GST) has been used to inhibit H1N1 virus infection, providing complementary evidence to antibody blocking experiments .
When feasible, validate blocking results using genetic approaches (siRNA knockdown or CRISPR-mediated deletion of GRM2).
Cytotoxicity Monitoring:
Assess potential antibody-induced cytotoxicity at the concentrations used for blocking experiments to ensure that observed effects are not due to cell death .
Careful attention to these considerations helps ensure that functional blocking experiments with GRM2 antibodies yield reliable and interpretable results.
GRM2 has been implicated in hippocampus-dependent memory and psychiatric disorders like schizophrenia. When investigating these connections, researchers can employ several antibody-based approaches:
Spatial Expression Mapping:
Use immunohistochemistry with GRM2 antibodies to map expression patterns across hippocampal subregions (CA1, CA3, dentate gyrus) and compare expression between control and disease models .
Co-label with markers for specific neuronal populations to determine which cell types express GRM2. Studies have used PROX1 (for dentate gyrus granule cells), DCX (for immature neurons), and MAP2 (for mature neurons) in conjunction with GRM2 antibodies .
Activity-Dependent Regulation:
Employ phospho-specific antibodies to track activation states of downstream signaling molecules including ERK1/2, MEK, CREB, and PKA-C, which are modulated by GRM2 activity .
Western blot analysis of these signaling components can reveal how GRM2 activation influences memory-related signaling cascades under different behavioral conditions .
Experimental Design for Memory Studies:
Combine behavioral paradigms with immunohistochemistry to correlate GRM2 expression/localization with memory performance.
When designing such experiments, consider that GRM2/3 knockout mice show a fractionation of hippocampus-dependent memory based on appetitive-aversive context. These mice are impaired on appetitively motivated spatial memory tasks but perform normally on aversively motivated tasks, suggesting task-specific roles for GRM2 .
Methodological Approach Table:
| Research Question | Antibody Technique | Complementary Methods | Key Controls |
|---|---|---|---|
| Cell-type specificity | Multi-color IHC with cell markers | Single-cell RNA-seq | GRM2 knockout tissue |
| Activity-dependent changes | Phospho-protein Western blots | Pharmacological manipulation | Vehicle-treated controls |
| Subcellular localization | Immunoelectron microscopy | Subcellular fractionation | Pre-absorption controls |
| Receptor trafficking | Surface biotinylation + GRM2 IP | Live-cell imaging | Temperature controls (4°C vs 37°C) |
This multi-faceted approach can provide comprehensive insights into GRM2's role in hippocampal function and related disorders.
Recent research has revealed an unexpected role for GRM2 as an endocytic receptor for influenza virus entry . For researchers investigating this novel function, several antibody-based approaches are valuable:
Viral Attachment and Entry Assays:
Use fluorescently-labeled virus particles in conjunction with anti-GRM2 antibodies to visualize co-localization at the cell surface during viral attachment.
Employ blocking antibodies targeting the GRM2 ectodomain to inhibit viral entry in dose-response experiments. Previous studies have shown that monoclonal antibodies against GRM2 can inhibit H1N1 virus infection in a dose-dependent manner .
Co-immunoprecipitation Studies:
Perform co-immunoprecipitation with anti-GRM2 antibodies to pull down viral hemagglutinin (HA) protein and demonstrate direct protein-protein interaction.
For robust demonstration of interaction specificity, reciprocal co-IP with anti-HA antibodies should be performed to pull down GRM2.
Endocytosis and Trafficking Visualization:
Combine anti-GRM2 antibodies with markers of clathrin-mediated endocytosis (CME) to track receptor internalization during viral entry.
Use pulse-chase experiments with surface-bound anti-GRM2 antibodies to follow receptor internalization kinetics in the presence and absence of virus.
Experimental Controls and Validations:
Include competition assays with soluble GRM2 ectodomain (e.g., mGluR2-GST) to demonstrate specificity of the interaction .
Verify that anti-GRM2 antibodies do not affect cell viability at concentrations used for blocking experiments .
Perform parallel experiments in GRM2-knockout or GRM2-depleted cells to confirm the specificity of observed effects.
Quantitative Assessment Methods:
For quantifying infection inhibition, researchers have successfully employed infectivity neutralization assays with purified GRM2 ectodomain or anti-GRM2 antibodies .
Flow cytometry can be used to quantify the percentage of infected cells under different conditions of GRM2 blockade or knockdown.
These methodological approaches provide a comprehensive framework for investigating GRM2's novel role in viral infection processes.
GRM2 has been implicated in the functional integration of adult-born dentate gyrus cells (DGCs) . When studying its developmental roles, researchers should consider these methodological approaches:
Temporal Expression Analysis:
Use GRM2 antibodies in conjunction with developmental markers to track expression changes during neuronal maturation.
In studies of adult-born DGCs, researchers have combined GRM2 immunostaining with doublecortin (DCX, immature neurons) and MAP2 (mature neurons) to correlate GRM2 expression with maturation state .
Genetic Manipulation Coupled with Antibody Detection:
Employ retroviral or lentiviral vectors expressing short hairpin RNAs (shRNAs) against GRM2 to knockdown expression in specific neuronal populations.
Validate knockdown efficiency using anti-GRM2 antibodies in Western blot analysis, with quantification relative to internal controls like β-Tubulin .
Pathway Analysis Through Phospho-specific Antibodies:
Monitor activation of downstream signaling cascades using phospho-specific antibodies for key components like ERK1/2, MEK, CREB, and PKA-C.
Western blot analysis with these antibodies can reveal how GRM2 modulates signaling during different developmental stages .
Subcellular Localization During Development:
Track GRM2 localization changes during neuronal maturation using high-resolution microscopy with anti-GRM2 antibodies.
Co-stain with synaptic markers like PSD95 and Homer1 to examine GRM2 recruitment to developing synapses .
Methodological Workflow for Developmental Studies:
Baseline Characterization:
Quantify normal GRM2 expression trajectory during development using time-course immunostaining and Western blot analysis.
Functional Manipulation:
Implement knockdown or overexpression strategies targeting GRM2.
Validate manipulation efficiency with anti-GRM2 antibodies.
Consequence Assessment:
Examine morphological changes using structural markers and standard immunohistochemistry.
Analyze signaling alterations with phospho-specific antibodies.
Evaluate functional outcomes through electrophysiology or behavioral assays.
Rescue Experiments:
Attempt to rescue phenotypes through reintroduction of GRM2 or downstream effectors.
Validate the specificity of rescue with appropriate antibody detection.
This systematic approach enables comprehensive investigation of GRM2's developmental functions while ensuring appropriate validation at each experimental stage.
Non-specific binding is a common challenge when working with GRM2 antibodies, particularly in neural tissues with complex protein expression profiles. To minimize these issues:
Antibody Selection Strategies:
Choose antibodies validated against GRM2 knockout tissues whenever possible.
Review BLAST analysis data for the antibody's immunogen to ensure minimal homology with other proteins. Some commercial GRM2 antibodies show homology with GRM3 (up to 50%) which can lead to cross-reactivity .
Protocol Optimizations for Western Blot:
Increase blocking stringency using 5% non-fat milk or 5% BSA in TBS-T for at least 1 hour at room temperature.
Implement additional washing steps (5-6 washes of 10 minutes each) with TBS-T after primary and secondary antibody incubations.
Titrate primary antibody concentration; for example, dilutions ranging from 1:500 to 1:3000 have been used successfully for different GRM2 antibodies .
Immunohistochemistry Background Reduction:
For highly autofluorescent tissues like brain, consider using Sudan Black B (0.1-0.3%) treatment after secondary antibody incubation to quench autofluorescence.
Extend blocking time to 2 hours using a combination of 5% normal serum from the secondary antibody host species plus 2% BSA.
Employ antigen retrieval optimization: test both citrate buffer (pH 6.0) and EDTA buffer (pH 9.0) to determine which yields better signal-to-noise ratio.
Validation Controls Table:
| Control Type | Implementation | Purpose |
|---|---|---|
| Negative control | Primary antibody omission | Detects non-specific secondary antibody binding |
| Absorption control | Pre-incubate antibody with immunizing peptide | Confirms epitope specificity |
| Genetic control | GRM2 knockdown/knockout tissue | Gold standard for specificity validation |
| Cross-reactivity control | Test on tissues with known GRM2/GRM3 expression | Distinguishes between related receptors |
By systematically implementing these strategies, researchers can significantly improve signal specificity when working with GRM2 antibodies.
Validating antibody specificity is crucial for generating reliable data with GRM2 antibodies. A comprehensive validation approach includes:
Genetic Validation Approaches:
Use of GRM2 knockout animals or cells represents the gold standard for specificity validation.
When knockout models aren't available, siRNA or shRNA-mediated knockdown of GRM2 can provide valuable validation data. For example, researchers have used shRNA against different regions of mouse Grm2 to validate antibody specificity .
Orthogonal Detection Methods:
Complement antibody-based detection with mRNA analysis (qPCR, RNA-seq, or in situ hybridization) to correlate protein detection with transcript presence.
When discrepancies arise between antibody detection and mRNA expression, additional validation steps are essential.
Multiple Antibody Verification:
Use multiple antibodies targeting different epitopes of GRM2 and compare their detection patterns.
Consistent results across antibodies targeting distinct regions (N-terminal vs. C-terminal) provide stronger evidence for specificity.
Peptide Competition Assays:
Pre-incubate the antibody with excess immunizing peptide before application to the sample.
Specific signal should be significantly reduced or eliminated after peptide competition.
Expression System Validation:
Test antibody on cells transfected with GRM2 expression vectors versus empty vector controls.
For quantitative validation, create a dilution series of recombinant GRM2 to establish detection limits and linear range.
Cross-reactivity Assessment:
Test on tissues or cells expressing related receptors (particularly GRM3) to evaluate potential cross-reactivity.
BLAST analysis of the immunizing peptide sequence can predict potential cross-reactivity. Some commercial GRM2 antibodies show up to 50% homology with GRM3 .
| Validation Method | Weight | Implementation |
|---|---|---|
| Genetic model | 5 | Knockout/knockdown verification |
| Orthogonal method | 4 | mRNA/protein correlation |
| Multiple antibodies | 3 | Consistent results with different epitopes |
| Peptide competition | 3 | Signal elimination with immunizing peptide |
| Expression system | 3 | Recombinant protein detection |
| Cross-reactivity | 2 | Testing on related proteins |
A cumulative score ≥10 would indicate well-validated antibody specificity for GRM2 detection.
Understanding GRM2's interactions with other proteins is crucial for deciphering its signaling mechanisms and functional roles. Several antibody-based approaches can effectively capture these interactions:
Co-immunoprecipitation Strategies:
For optimal pull-down of membrane-bound GRM2 and its interacting partners, use mild detergents like 1% CHAPS or 0.5% NP-40 to preserve protein-protein interactions.
Pre-clearing lysates with protein A/G beads can reduce non-specific binding.
Cross-linking approaches (e.g., DSP or formaldehyde) before lysis can stabilize transient interactions that might otherwise be lost during solubilization.
Proximity Ligation Assay (PLA):
PLA provides high sensitivity for detecting protein-protein interactions in situ with resolution below 40 nm.
This technique has advantages for detecting interactions between GRM2 and other membrane proteins or cytoskeletal components in their native cellular context.
Careful optimization of primary antibody dilutions is essential, typically using more dilute concentrations than standard immunofluorescence.
Bimolecular Fluorescence Complementation (BiFC):
When combined with antibody detection, BiFC can validate direct protein interactions while simultaneously localizing the complexes within cellular compartments.
Anti-GRM2 antibodies can be used to confirm the expression and proper localization of GRM2-fusion constructs.
FRET Analysis with Antibody Validation:
FRET microscopy using fluorophore-conjugated antibodies can detect close proximity between GRM2 and candidate interacting proteins.
This approach requires careful selection of non-competing antibodies and appropriate fluorophore pairs.
Interactome Analysis Workflow:
Initial screening: Use co-IP combined with mass spectrometry to identify potential interacting partners.
Validation: Confirm key interactions through reciprocal co-IP and Western blotting.
Localization: Employ PLA or confocal microscopy with co-labeling to determine where interactions occur.
Functional assessment: Test interaction significance through mutagenesis of binding domains followed by co-IP verification.
Recent Applications:
Recent studies have used these techniques to investigate interactions between GRM2 and key signaling molecules. For example, researchers have identified interactions between GRM2 and components of the MAPK pathway, including ERK1/2, MEK, c-Raf, and b-Raf, providing insights into how GRM2 regulates neural development .
By applying these methodological approaches, researchers can comprehensively map GRM2's interactome and gain insights into its diverse functional roles.
Deep learning and computational approaches are increasingly valuable for optimizing antibody research, particularly for challenging targets like GRM2:
Epitope Prediction and Antibody Design:
Recent advances in protein structural prediction (e.g., AlphaFold2) can generate high-confidence models of GRM2's structure, identifying optimal epitopes that maximize specificity and accessibility.
These computational predictions can guide the design of synthetic peptide immunogens targeting regions with minimal homology to related proteins like GRM3.
Machine learning algorithms can analyze existing antibody sequence-specificity relationships to predict which antibody sequences might offer improved specificity for GRM2 over related proteins.
Cross-Reactivity Prediction:
Computational tools can systematically compare potential epitopes across the proteome to identify possible cross-reactivity.
Similar to approaches used in antibody studies for SARS-CoV-2 spike protein, deep learning models can be trained to differentiate sequences of antibodies targeting different but related proteins .
These models could help predict whether a given antibody sequence is likely to recognize GRM2 specifically versus related metabotropic glutamate receptors.
Image Analysis Enhancement:
Deep learning algorithms can improve the specificity of GRM2 detection in complex tissues by:
Reducing background and autofluorescence in immunofluorescence images
Automatically distinguishing specific from non-specific staining patterns
Quantifying co-localization with other proteins with greater precision than traditional methods
Implementation Table for Computational Approaches:
| Computational Method | Application | Implementation Complexity | Potential Impact |
|---|---|---|---|
| Structural prediction | Epitope selection | Medium | High - enables rational antibody design |
| Sequence analysis | Cross-reactivity assessment | Low | Medium - improves antibody selection |
| Deep learning classification | Antibody specificity prediction | High | High - could revolutionize validation |
| AI image analysis | Immunostaining interpretation | Medium | Medium - enhances data quality |
| Molecular dynamics | Antibody-antigen interaction | High | Medium - refines binding predictions |
Practical Research Application:
In a proof-of-concept study for SARS-CoV-2 antibodies, researchers demonstrated that deep learning models could differentiate antibody sequences targeting spike protein versus influenza hemagglutinin with high accuracy (89%) . Similar approaches could be applied to develop models for predicting GRM2 antibody specificity, potentially accelerating research by helping select optimal antibodies or designing new ones with enhanced specificity.
These computational approaches represent the cutting edge of antibody research and offer significant potential for advancing GRM2 studies.
GRM2 undergoes various post-translational modifications (PTMs), including glycosylation , which significantly affect its function, localization, and interactions. Studying these PTMs requires specialized antibody-based approaches:
Glycosylation Analysis:
Use lectins in conjunction with anti-GRM2 antibodies to detect and characterize specific glycan structures on GRM2.
Enzymatic deglycosylation (PNGase F for N-linked or O-glycosidase for O-linked glycans) followed by Western blot analysis with anti-GRM2 antibodies can reveal the extent and sites of glycosylation.
Mobility shift analysis comparing native and deglycosylated GRM2 can provide insights into the contribution of glycans to the protein's apparent molecular weight.
Phosphorylation Detection:
Employ phospho-specific antibodies targeting known or predicted phosphorylation sites on GRM2.
Enhance detection sensitivity by first immunoprecipitating GRM2 using general anti-GRM2 antibodies, then probing with phospho-specific antibodies.
Validation should include treatments with phosphatase inhibitors (e.g., okadaic acid) to preserve phosphorylation states and phosphatase treatments as negative controls.
Ubiquitination and SUMOylation Analysis:
Use a dual immunoprecipitation approach: first pull down GRM2, then probe for ubiquitin or SUMO modifications.
Alternatively, immunoprecipitate ubiquitinated or SUMOylated proteins and probe for GRM2.
Proteasome inhibitors (e.g., MG132) can be used to enhance detection of ubiquitinated species that might otherwise be rapidly degraded.
Mass Spectrometry Integration:
Combine immunoprecipitation using anti-GRM2 antibodies with mass spectrometry analysis to identify and map PTMs comprehensively.
This approach can reveal unexpected modifications and provide precise localization of modification sites.
PTM Site-Specific Antibody Development:
For critical PTM sites, consider developing custom antibodies that specifically recognize the modified form of GRM2.
Validate these antibodies using site-directed mutagenesis of the modification site as a negative control.
Experimental Design Considerations:
| PTM Type | Recommended Detection Method | Key Controls | Physiological Significance Assessment |
|---|---|---|---|
| Glycosylation | Lectin blotting + GRM2 antibodies | Deglycosylation enzymes | Compare receptor function before/after deglycosylation |
| Phosphorylation | Phospho-specific antibodies | Phosphatase treatment | Analyze signaling changes under stimulation/inhibition |
| Ubiquitination | Co-IP with ubiquitin antibodies | Proteasome inhibitors | Measure protein stability and turnover rates |
| Palmitoylation | Click chemistry + GRM2 antibodies | Hydroxylamine treatment | Assess membrane localization dependencies |
By employing these specialized approaches, researchers can gain detailed insights into how PTMs regulate GRM2 function, potentially revealing new therapeutic targets or diagnostic markers.