GRAP2 (GRB2-related adaptor protein 2) is a member of the GRB2/Sem5/Drk family functioning as an adaptor protein involved in leukocyte-specific protein-tyrosine kinase signaling. Its structural organization includes a central SH2 domain flanked by two SH3 domains, with a unique 120-amino acid glutamine/proline rich sequence not found in GRB2 or GRAP .
GRAP2 is primarily expressed in lymphoid tissues and hematopoietic cells, particularly T cells, where it plays a pivotal role during early events of T cell signal transduction . Mechanistically, GRAP2:
Interacts with SLP-76 to regulate NF-AT (nuclear factor of activated T cells) activation
Forms signaling complexes with HPK1 to mediate the JNK signaling pathway
Recruits adaptor protein SLP-76 and its associated molecules (Vav, Nck, Itk, ADAP) to the transmembrane adaptor protein LAT
Interacts with M-CSF receptor and the activated T cell co-stimulatory receptor CD28
Associates with other signaling proteins including Gab2, HPK1, and Cbl
Recent studies have identified GRAP2 as a potential prognostic biomarker in certain cancers, making it increasingly relevant to both immunological and oncological research .
GRAP2 antibodies have been validated for multiple applications, with varying effectiveness depending on the specific antibody clone and experimental conditions:
For optimal results:
Western blotting is most widely validated and shows consistent results across different antibodies
Flow cytometry requires specific conjugated antibodies for best results
IHC applications benefit from antigen retrieval optimization
Cross-validate findings using at least two different detection methods
Validating antibody specificity is crucial for obtaining reliable results. For GRAP2 antibodies, several validation strategies are recommended:
Compare protein expression using antibody-based and antibody-independent methods:
Correlate antibody detection with mRNA expression data from RT-PCR or RNA-seq
Compare results with mass spectrometry-based protein detection
Use multiple antibodies targeting different epitopes of GRAP2
Compare staining patterns between antibodies recognizing different regions (e.g., N-terminal vs. C-terminal)
Antibodies targeting different domains (SH2 vs. SH3) should show similar expression patterns
Use CRISPR/Cas9 knockout or siRNA knockdown of GRAP2
Absence of signal in knockout samples confirms specificity
Overexpress tagged GRAP2 in cell lines with low endogenous expression
Confirm co-localization of anti-GRAP2 antibody with tag-specific antibodies
As described in the Enhanced Validation methodology, compare IHC staining pattern with RNA expression levels to determine consistency scores :
High consistency: Strong correlation between antibody staining and RNA expression
Medium consistency: General agreement with some discrepancies
Low consistency: Limited correlation requiring additional validation
The detection of GRAP2 requires tailored approaches for different biological samples:
Flow Cytometry (Preferred): Use PE or APC-conjugated anti-GRAP2 antibodies for intracellular staining
Western Blot:
IHC on Paraffin Sections:
Pre-treatment: Heat-induced epitope retrieval in citrate buffer (pH 6.0)
Primary antibody incubation: Overnight at 4°C
Detection: HRP-polymer and DAB visualization
Counterstain: Hematoxylin
GRAP2 expression is generally lower in non-lymphoid tissues, requiring:
Enhanced signal amplification techniques
Longer primary antibody incubation times
More sensitive detection systems (e.g., TSA amplification)
For lung adenocarcinoma specifically, IHC has confirmed lower GRAP2 protein expression in tumor tissues compared to adjacent normal tissues .
Research has revealed significant relationships between GRAP2 expression and immune infiltration in cancer:
GRAP2 expression positively correlates with infiltration levels of multiple immune cell types in tumors
A hub gene set of 91 genes coexpressed with GRAP2 was found to be closely related to immune response in LUAD
GRAP2 expression positively correlates with multiple immune markers, chemokines, chemokine receptors, and MHC molecules
Analyze GRAP2 expression using RNA-seq or microarray data from cancer databases (TCGA, GEO)
Quantify immune cell infiltration using computational approaches (TIMER, CIBERSORT)
Perform correlation analysis between GRAP2 expression and immune infiltration metrics
Validate findings using multiplex immunofluorescence or immunohistochemistry
Conduct functional studies to elucidate mechanistic relationships
These findings suggest GRAP2 could serve as a biomarker for assessing both prognosis and immune infiltration levels in certain cancers .
Detecting post-translational modifications (PTMs) of GRAP2 presents several technical challenges:
GRAP2 can undergo phosphorylation at specific tyrosine residues
PTMs may alter protein-protein interactions, particularly with binding partners like SLP-76 and LAT
Modified GRAP2 can generate autoantibodies in certain conditions
Limited PTM-specific antibodies: Few commercial antibodies specifically target modified forms of GRAP2
Low abundance of modified forms: PTM-bearing GRAP2 may represent a small fraction of total GRAP2
Labile modifications: Some PTMs may be lost during sample preparation
Background interference: Cross-reactivity with other modified proteins may occur
Phospho-specific antibodies: When available, use antibodies specifically targeting phosphorylated residues
Mass spectrometry:
Immunoprecipitate GRAP2 followed by LC-MS/MS analysis
Use neutral loss scanning to detect phosphorylation events
Apply electron transfer dissociation (ETD) for improved PTM site identification
Phos-tag SDS-PAGE: For detecting phosphorylated GRAP2 variants with mobility shift
2D gel electrophoresis: To separate GRAP2 isoforms with different PTMs
Use phosphatase treatment as negative control for phosphorylation
Compare stimulated vs. unstimulated cells to identify inducible modifications
Express mutant GRAP2 (with modified PTM sites) as controls
Proper storage and handling of GRAP2 antibodies is critical for maintaining their activity and specificity:
Most GRAP2 antibodies are supplied in:
Avoid repeated freeze/thaw cycles (more than 3-5 cycles significantly reduces activity)
For -20°C storage of larger volumes (>20μL), aliquoting is recommended
Small volume antibodies (20μL) containing 0.1% BSA can be stored without aliquoting
Allow antibodies to equilibrate to room temperature before opening
Briefly centrifuge before use to collect solution at the bottom of the tube
For conjugated antibodies, protect from light during all handling steps
Diluted antibody working solutions are typically stable for up to 7 days at 2-8°C
For longer-term storage of working dilutions, add carrier protein (e.g., 1% BSA)
Sodium azide (0.02%) can be added to prevent microbial growth, but may interfere with some applications (e.g., HRP detection systems)
Various GRAP2 antibody clones show different performance characteristics across applications:
Specificity: Monoclonal antibodies generally show higher specificity but recognize single epitopes
Sensitivity: Polyclonal antibodies often provide higher sensitivity by binding multiple epitopes
Background: Monoclonal antibodies typically generate lower background in immunohistochemistry
Batch consistency: Monoclonal antibodies offer better lot-to-lot consistency
Species cross-reactivity: Polyclonal antibodies often have broader species cross-reactivity
For critical experiments, validation using more than one antibody clone is recommended to confirm findings .
GRAP2 has emerging potential as a cancer biomarker, with several methodological approaches for investigation:
GRAP2 has been identified as a prognostic biomarker in lung adenocarcinoma and cervical cancer
Lower GRAP2 expression correlates with poorer prognosis in LUAD patients
Tissue Microarray (TMA) Analysis:
Collect tumor samples and matched normal tissues
Prepare TMA blocks and perform IHC using validated GRAP2 antibodies
Score expression levels based on staining intensity and percentage of positive cells
Correlate with clinical parameters and survival data
Transcriptomic Analysis:
Protein-Protein Interaction Studies:
Perform co-immunoprecipitation using GRAP2 antibodies
Identify interaction partners in normal vs. cancer tissues
Investigate altered signaling pathways in cancer context
Immune Infiltration Correlation:
Use multiplexed IHC to simultaneously detect GRAP2 and immune cell markers
Quantify immune cell types in relation to GRAP2 expression levels
Correlate findings with response to immunotherapy where applicable
Use antibodies with demonstrated specificity in cancer tissues
Include positive controls (lymphoid tissues) and negative controls
Consider heterogeneity within tumors by analyzing multiple regions
Validate findings across independent patient cohorts
Combine with other established biomarkers for improved prognostic value
GRAP2 has been identified as a binding partner of RET receptor tyrosine kinase, with implications for neuroendocrine tumors:
Co-immunoprecipitation:
In vitro Binding Assays:
Functional Studies:
GRAP2 is expressed in neuroendocrine tumors and cell lines bearing mutated RET
Endogenous RET and GRAP2 co-immunoprecipitate from medullary thyroid carcinoma cell lines
GRAP2 inhibits RET-induced NF-κB activation
GRAP2 overexpression reduces focus formation induced by oncogenic RET
Include proper controls for immunoprecipitation (isotype control antibodies)
Validate interactions using multiple antibodies targeting different epitopes
Consider the cell type-specific context, as GRAP2 is predominantly expressed in hematopoietic cells
Examine the effects of RET activation (ligand stimulation or oncogenic mutations) on GRAP2 binding
Investigate the role of GRAP2 phosphorylation in mediating RET interactions
These findings suggest GRAP2 may play a tissue-specific role as an inhibitor of RET mitogenic signaling, extending its known functions beyond hematopoietic cell signaling .