EVI2A (ecotropic viral integration site 2A) is a membrane-localized protein with a length of 236 amino acid residues and a molecular weight of approximately 26.2 kDa in humans. Functionally, EVI2A is believed to form complexes either with itself or with other proteins within the cellular membrane, serving as a component of a cell-surface receptor system. The protein undergoes post-translational modifications, particularly glycosylation, which may be critical for its proper functioning . Current research suggests its involvement in cellular signaling pathways, though the complete spectrum of its biological activities remains under investigation .
EVI2A antibodies are utilized across several experimental platforms in molecular and cellular biology research:
| Application | Description | Common Protocols |
|---|---|---|
| Western Blot (WB) | Most widely used for protein detection and quantification | Typically using 20-50 μg of total protein lysate with 1:500-1:2000 antibody dilution |
| ELISA | Quantitative detection of EVI2A in solution | Usually at 1:1000-1:5000 dilution depending on antibody sensitivity |
| Immunohistochemistry (IHC) | Tissue localization studies | Often requires 1:50-1:200 dilution with antigen retrieval methods |
| Immunofluorescence (IF) | Subcellular localization | Generally at 1:100-1:500 dilution with appropriate secondary antibodies |
| Flow Cytometry (FACS) | Cell surface expression analysis | Typically using 1μg antibody per 10^6 cells in suspension |
The selection of application should be guided by experimental questions and available validation data for specific antibody products .
Researchers can access several categories of EVI2A antibodies, each with distinct properties:
| Antibody Type | Characteristics | Best Applications |
|---|---|---|
| Polyclonal | Recognizes multiple epitopes; higher sensitivity but potential for cross-reactivity | Western blot, ELISA, preliminary experiments |
| Monoclonal | Recognizes single epitope; higher specificity | Flow cytometry, quantitative applications requiring reproducibility |
| Unconjugated | Requires secondary detection method | Flexible for various applications with amplification steps |
| Conjugated | Direct detection via fluorophores, enzymes, or tags | Flow cytometry, direct detection methods |
Additionally, antibodies are available with reactivity to EVI2A from various species including human, mouse, rat, cow, horse, pig, bat, and monkey, allowing for comparative studies across model organisms .
Expression correlation data from studies examining tumor microenvironment interactions show:
| Cancer Type | Expression Pattern | Clinical Correlation | Statistical Significance |
|---|---|---|---|
| KIRC | Upregulated | Poor prognosis, advanced stage | p < 0.001, AUC = 0.906 |
| Esophageal Cancer | Environmental response gene | Prognostic indicator | Significant correlation with survival metrics |
| Peripheral Nerve Sheath Tumors | Associated with NF1 deficiency | Increased susceptibility | Correlation in NF1-deficient models |
Methodologically, researchers investigating EVI2A in pathological contexts should consider combining transcriptomic analysis (RNA-seq) with protein-level studies (IHC, Western blot) to establish consistent patterns across biological scales .
Research indicates that EVI2A expression is regulated through multiple mechanisms, with epigenetic modification playing a particularly important role. Current evidence identifies EVI2A as a methylation-related gene in KIRC and other cancers, suggesting that DNA methylation status significantly impacts its transcriptional activity .
For researchers investigating regulatory mechanisms, the following methodological approaches are recommended:
Methylation-specific PCR and bisulfite sequencing to assess promoter methylation status
Chromatin immunoprecipitation (ChIP) to identify transcription factor binding
Reporter gene assays to evaluate promoter activity under different conditions
Treatment with epigenetic modifiers (e.g., 5-azacytidine) to experimentally manipulate methylation status
Additionally, researchers should consider examining microRNA regulation and histone modifications as complementary regulatory mechanisms that may interact with DNA methylation to fine-tune EVI2A expression in different cellular contexts .
Detection of post-translational modifications (PTMs) of EVI2A presents special challenges due to the protein's membrane localization and glycosylation status. Current evidence indicates that glycosylation is a significant PTM for EVI2A , potentially affecting antibody recognition and experimental outcomes.
For optimal detection of glycosylated EVI2A:
| Approach | Methodology | Considerations |
|---|---|---|
| Deglycosylation | Treatment with PNGase F or Endo H prior to Western blot | May reveal multiple bands reflecting differentially glycosylated isoforms |
| Lectin affinity | Combined with immunoprecipitation | Can identify specific glycan structures on EVI2A |
| Specialized extraction | Membrane protein extraction buffers with glycoprotein solubilizers | Improves recovery of intact glycosylated forms |
| Mass spectrometry | Glycoproteomics analysis | Provides detailed structural information about glycan composition |
Researchers should select antibodies specifically validated for detecting the protein regardless of glycosylation state or choose epitopes known to be unaffected by this modification .
Western blotting for EVI2A requires specific optimization due to its membrane localization and post-translational modifications. Recommended protocol adjustments include:
Sample preparation:
Use membrane protein extraction buffers containing 1-2% non-ionic detergents (Triton X-100 or NP-40)
Include protease inhibitors and maintain samples at 4°C throughout processing
Consider using specialized glycoprotein solubilization buffers if targeting glycosylated forms
Gel electrophoresis parameters:
10-12% polyacrylamide gels provide optimal resolution for the 26.2 kDa protein
Expected migration pattern: 26-30 kDa (unmodified) or 35-45 kDa (glycosylated forms)
Include positive control lysates from tissues with known EVI2A expression
Transfer and detection:
Semi-dry transfer systems at 15-20V for 30-45 minutes typically yield efficient transfer
Block with 5% non-fat milk or BSA in TBST
Primary antibody incubation: overnight at 4°C at dilutions between 1:500-1:2000
HRP-conjugated secondary antibodies at 1:5000-1:10000 for 1 hour at room temperature
These experimental conditions should be further optimized based on specific antibody characteristics and sample types .
Immunoprecipitation (IP) of EVI2A presents unique challenges due to its membrane localization. The following methodological approach is recommended:
Cell lysis optimization:
Use NP-40 or digitonin-based lysis buffers (0.5-1%) to solubilize membrane proteins while preserving protein-protein interactions
Include phosphatase inhibitors if studying phosphorylation-dependent interactions
Perform lysis at 4°C with gentle agitation for 30-60 minutes
Antibody selection considerations:
Choose antibodies raised against regions not involved in protein-protein interactions
Validate antibody efficiency in preliminary Western blot experiments
For co-IP studies, epitope-tagged EVI2A constructs may provide higher specificity
Protocol optimization:
Pre-clear lysates with Protein A/G beads for 1 hour at 4°C
Use 2-5 μg antibody per 500 μg of total protein
Extend incubation time to overnight at 4°C with gentle rotation
Include appropriate negative controls (isotype control antibodies)
Analysis of interacting partners:
Mass spectrometry-based identification of co-precipitated proteins
Validation of key interactions through reciprocal co-IP and proximity ligation assays
This approach maximizes the likelihood of preserving physiologically relevant interactions while minimizing non-specific binding .
Rigorous experimental design for EVI2A antibody applications should include several critical controls:
| Control Type | Implementation | Purpose |
|---|---|---|
| Positive Control | Lysates from tissues with confirmed high EVI2A expression (e.g., specific immune cells) | Verifies antibody functionality and expected band pattern |
| Negative Control | Tissues known to lack EVI2A expression or EVI2A-knockout samples | Establishes specificity and identifies non-specific binding |
| Loading Control | Probing for housekeeping proteins (β-actin, GAPDH) or total protein staining | Ensures equal loading across samples for quantitative comparisons |
| Antibody Specificity Control | Pre-absorption with immunizing peptide | Confirms epitope-specific binding |
| Secondary Antibody Control | Omission of primary antibody | Identifies non-specific binding of secondary antibody |
| siRNA/shRNA Validation | Comparison with EVI2A-knockdown samples | Confirms signal specificity through targeted reduction |
For immunohistochemistry or immunofluorescence, additional tissue-specific controls and counterstaining should be employed to verify subcellular localization patterns .
Non-specific binding is a common challenge when working with membrane protein antibodies. For EVI2A antibodies, consider these methodological solutions:
Antibody selection refinement:
Choose monoclonal antibodies for higher specificity when possible
Review validation data showing specificity across multiple applications
Select antibodies targeting unique regions of EVI2A with minimal homology to related proteins
Protocol optimization:
Increase blocking stringency (5% BSA or 5-10% normal serum from secondary antibody host species)
Add 0.1-0.3% Triton X-100 to blocking and antibody dilution buffers
Extend blocking time to 2 hours at room temperature
Perform additional washing steps (5-6 washes of 10 minutes each)
Titrate primary antibody concentration through dilution series experiments
Sample preparation improvements:
Pre-absorb antibodies with proteins from species of non-interest
Use freshly prepared samples to reduce degradation products
Consider antigen retrieval optimization for fixed samples
Data interpretation:
Compare multiple antibodies targeting different EVI2A epitopes
Correlate protein detection with mRNA expression data
Use genetic manipulation (siRNA, CRISPR) to validate specificity
These approaches should significantly reduce non-specific binding while preserving authentic EVI2A signal .
EVI2A detection may be challenging in cell types with naturally low expression levels. Implementation of signal amplification strategies can help overcome this limitation:
Enhanced protein extraction:
Scale up starting material (3-5x typical amounts)
Implement subcellular fractionation to concentrate membrane proteins
Use specialized extraction buffers optimized for glycosylated membrane proteins
Signal amplification techniques:
Employ tyramide signal amplification (TSA) for immunohistochemistry/immunofluorescence
Use high-sensitivity ECL substrates for Western blotting
Consider biotin-streptavidin amplification systems
Implement polymeric detection systems for IHC
Sample enrichment prior to analysis:
Perform immunoprecipitation before Western blotting
Use lectin-based enrichment for glycosylated forms
Consider using more sensitive detection methods like droplet digital PCR at the mRNA level to correlate with protein findings
Innovative detection platforms:
Single molecule array (Simoa) technology for ultra-sensitive protein detection
Proximity extension assay (PEA) for detecting EVI2A in complex samples
Mass cytometry (CyTOF) for simultaneous detection of multiple parameters
These approaches can lower detection limits by 10-100 fold compared to standard methods .
Sample preparation significantly impacts the detection of membrane-associated proteins like EVI2A. The following methodological comparison may guide researchers:
| Preparation Method | Advantages | Limitations | Best Applications |
|---|---|---|---|
| RIPA Buffer Extraction | Good protein yield, compatible with many applications | May disrupt some epitopes, especially in transmembrane regions | Western blot, ELISA |
| NP-40/Triton X-100 (0.5-1%) | Preserves most protein-protein interactions | Lower extraction efficiency for strongly membrane-associated proteins | Co-immunoprecipitation, activity assays |
| Digitonin (0.01-0.1%) | Very gentle, maintains membrane protein complexes | Limited solubilization, requires optimization | Blue native PAGE, structure studies |
| Membrane Fractionation | Enriches for EVI2A, reduces background | Time-consuming, potential for artifacts during preparation | Studies focused specifically on membrane proteins |
| Formaldehyde Fixation | Preserves tissue architecture | May mask epitopes, requiring antigen retrieval | IHC, IF on tissue sections |
| Methanol Fixation | Good for some membrane proteins | Can extract lipids, affecting membrane protein localization | IF of cultured cells |
For optimal results, researchers should test multiple preparation methods when establishing a new experimental system, particularly when working with novel antibodies or tissue sources .
EVI2A antibodies serve as valuable tools in cancer biomarker research, particularly given the protein's emerging role in multiple malignancies. Methodological approaches for biomarker application include:
Tissue microarray analysis:
Quantitative immunohistochemistry across tumor stages and grades
Correlation with patient outcomes using appropriate statistical methods
Multivariate analysis incorporating other established biomarkers
Liquid biopsy development:
Detection of EVI2A in circulation using highly sensitive immunoassays
Analysis of EVI2A in extracellular vesicles from patient serum
Correlation of EVI2A levels with disease progression
Multiparameter analysis:
Integration with other biomarkers in predictive models
Development of EVI2A-based prognostic panels
Combined analysis with genetic and epigenetic markers
Recent research specifically demonstrates EVI2A's significant diagnostic value in kidney renal clear cell carcinoma (KIRC) with an impressive AUC of 0.906, indicating excellent discriminatory power. Furthermore, its expression correlates with tumor stage, grade, and patient survival, suggesting potential utility in stratifying patients for treatment selection and monitoring disease progression .
EVI2A exhibits significant immunological associations that researchers can investigate using specialized antibody-based approaches:
Immune cell expression profiling:
Multicolor flow cytometry to map EVI2A expression across immune cell subsets
Single-cell analyses to identify specific populations with differential expression
Sequential immunophenotyping during immune cell activation/differentiation
Functional studies in immune contexts:
Investigation of EVI2A in immune synapse formation using high-resolution microscopy
Analysis of signaling pathway activation in EVI2A-expressing immune cells
Correlation with immune activation markers in different pathological states
Tumor microenvironment investigations:
Dual immunofluorescence for EVI2A and immune cell markers in tumor sections
Analysis of EVI2A expression in tumor-infiltrating lymphocytes
Correlation with immune checkpoint molecule expression
Current research indicates significant associations between EVI2A expression and specific immune cell populations, including T follicular helper cells, CD4+ memory T cells, and CD8+ T cells. Additionally, EVI2A expression correlates with tumor microenvironment scores and may indicate increased sensitivity to immune checkpoint inhibitors targeting PD-1/CTLA-4, suggesting potential implications for immunotherapy response prediction .
Advanced technological platforms are expanding the applications of EVI2A antibodies in research settings:
Single-cell technologies:
Mass cytometry (CyTOF) for high-dimensional protein profiling
Single-cell proteomics with antibody-based detection
Spatial transcriptomics combined with protein detection for tissue mapping
Advanced imaging approaches:
Super-resolution microscopy (STORM, PALM) for nanoscale localization
Expansion microscopy for enhanced visualization of subcellular structures
Live-cell imaging with antibody fragments for dynamic studies
Antibody engineering innovations:
Development of single-domain antibodies (nanobodies) for enhanced penetration and reduced size
BiTE (Bispecific T-cell Engager) antibodies for functional studies
Computationally designed antibodies with enhanced specificity and affinity
Therapeutic development platforms:
Antibody-drug conjugates targeting EVI2A in cancer models
CAR-T approaches utilizing EVI2A as a target in appropriate malignancies
Development of therapeutic antibodies against EVI2A for targeted therapy
Recent advances in computational antibody design, as demonstrated by the fine-tuned RFdiffusion network capable of designing de novo antibody variable heavy chains (VHH's), may soon allow for custom-designed anti-EVI2A antibodies with precisely engineered binding properties for specific research and therapeutic applications .