EYA1 antibodies are specialized immunological tools designed to detect and study the Eyes Absent Homolog 1 (EYA1) protein, a bifunctional molecule with tyrosine phosphatase activity and transcriptional coactivator roles . These antibodies are critical for investigating EYA1's involvement in organogenesis, cancer progression, and DNA repair mechanisms .
EYA1 antibodies are utilized across diverse experimental paradigms:
Melanoma: EYA1 overexpression correlates with tumor thickness, mitotic rate, and reduced survival. Knockdown reduces proliferation by 60% .
Neuroblastoma: High nuclear EYA1 levels associate with MYCN amplification and aggressive subtypes .
Glioma: EYA1 promotes tumor growth via BCL2L12 interaction; suppression extends survival in murine models .
Kidney Development: EYA1 specifies metanephric blastema; 20% protein levels suffice for ureteric bud induction .
Inner Ear Morphogenesis: Dosage-dependent effects on semicircular canal formation (e.g., cochlear agenesis at ≤21% EYA1 levels) .
EYA1 dephosphorylates histone H2AX at Tyr-142, facilitating MDC1-mediated DNA damage repair .
EYA1 (Eyes Absent Homolog 1) is a transcriptional coactivator and protein tyrosine phosphatase with a molecular weight of approximately 64.6 kilodaltons in humans . It functions as a critical developmental regulator and is also known by alternative names including BOP, BOR, BOS1, and OFC1 . EYA1 plays essential roles in organogenesis during embryonic development, particularly in eye, ear, and kidney formation. In pathological contexts, EYA1 has been identified as a regulator in cancer stem cells, notably in glioblastoma where it interacts with MYC to influence tumor cell proliferation, migration, and self-renewal . The dual functionality of EYA1 as both a phosphatase and transcriptional regulator makes it a complex and important target for research across developmental biology, cancer biology, and regenerative medicine fields.
Research-grade EYA1 antibodies are available in multiple formats with varying specificity and applications:
Antibody Type | Common Applications | Available Species Reactivity | Available Formats |
---|---|---|---|
Polyclonal | WB, ELISA, IHC, IF | Human, Mouse, Rat, Multiple species | Unconjugated, FITC-conjugated, Biotin-conjugated |
Monoclonal | WB, ELISA, ICC | Human, Mouse, Rhesus | Unconjugated |
Domain-specific | WB, IHC | Human, Mouse, Rat | Unconjugated |
Many suppliers offer antibodies targeting different epitopes of EYA1, including N-terminal, C-terminal, and middle region-specific antibodies . Some antibodies demonstrate cross-reactivity with other EYA family members (particularly EYA4), which should be considered when designing experiments requiring high specificity . Multiple species-reactive antibodies allow for comparative studies across model organisms, with human, mouse, and rat being the most commonly available reactivities.
Selection of the appropriate EYA1 antibody requires careful consideration of several experimental factors:
For co-immunoprecipitation studies examining EYA1-MYC interactions, antibodies validated for immunoprecipitation that target regions not involved in the interaction would be optimal .
For successful Western blotting with EYA1 antibodies, consider these protocol optimizations:
Sample preparation:
Gel selection and transfer:
Blocking and antibody incubation:
Block with 5% non-fat dry milk in TBST for 1 hour at room temperature
Dilute primary antibodies 1:500-1:2000 depending on manufacturer recommendations
Incubate with primary antibody overnight at 4°C
Use HRP-conjugated secondary antibodies at 1:5000-1:10000 dilution
Detection considerations:
The expected band size for human EYA1 is approximately 65 kDa
Verify specificity using positive controls (tissues/cells known to express EYA1)
Include negative controls such as knockdown samples when possible
When analyzing multiple EYA family members, perform sequential probing with appropriate stripping between antibodies to avoid cross-reactivity issues.
Co-immunoprecipitation (Co-IP) is valuable for studying EYA1 protein interactions, such as the EYA1-MYC axis in glioblastoma stem cells . For successful EYA1 Co-IP:
Cell preparation:
Pre-clearing and immunoprecipitation:
Washing and elution:
Wash immunoprecipitates 4-5 times with cold IP buffer
Elute bound proteins by boiling in SDS sample buffer for 5 minutes
Analysis and controls:
Include IgG control immunoprecipitations
Confirm interactions by reciprocal Co-IP (e.g., IP with anti-MYC and blot for EYA1)
Verify protein interactions by alternative methods (e.g., proximity ligation assay)
This experimental approach has successfully demonstrated EYA1-MYC binding in glioblastoma stem cells, revealing a potential regulatory mechanism .
EYA1 has been identified as preferentially expressed in glioblastoma stem cells (GSCs) compared to differentiated tumor progeny . To investigate EYA1's role in cancer stem cell biology:
Comparative expression analysis:
Use immunofluorescence with EYA1 antibodies to compare expression between stem-like and differentiated cancer cell populations
Co-stain with stem cell markers (CD133, SOX2, NESTIN) to correlate EYA1 expression with stemness
Functional studies with EYA1 knockdown:
Design lentiviral shRNA constructs targeting EYA1 (e.g., using validated sequences like TRCN0000369592, TRCN0000310704, or TRCN0000303462)
Confirm knockdown efficiency by Western blotting with EYA1 antibodies
Assess effects on stem cell properties: sphere formation, expression of stemness markers, multilineage differentiation potential
Regulatory network analysis:
In vivo validation:
Use EYA1 antibodies for immunohistochemical analysis of patient-derived xenografts
Compare EYA1 expression in tumor-initiating regions versus bulk tumor
This multifaceted approach can help elucidate EYA1's role in maintaining the cancer stem cell phenotype and identify potential therapeutic vulnerabilities, as demonstrated in glioblastoma research .
EYA1 possesses dual functionality as both a tyrosine phosphatase and transcriptional co-activator. Distinguishing between these functions requires specialized experimental approaches:
Phosphatase activity assays:
Immunoprecipitate EYA1 using specific antibodies and assess phosphatase activity with artificial substrates (p-nitrophenyl phosphate) or physiological substrates
Design domain-specific mutants (e.g., D327A mutation in the phosphatase domain) that selectively inhibit phosphatase function while preserving co-activator function
Use phosphatase-specific inhibitors to distinguish activity-dependent functions
Transcriptional co-activator function:
Perform ChIP assays using EYA1 antibodies to identify genomic binding sites
Use reporter gene assays with EYA1 binding sites to quantify transcriptional activation
Test co-immunoprecipitation with known transcriptional partners (SIX family proteins)
Dissecting dual functionality:
Use domain-specific antibodies that selectively recognize either the phosphatase domain or regions involved in transcriptional activation
Compare phospho-protein profiles using phospho-specific antibodies in cells with wild-type EYA1 versus phosphatase-dead mutants
Perform differential gene expression analysis after selectively inhibiting each function
Novel interaction studies:
These approaches collectively provide a comprehensive assessment of EYA1's dual functionality in experimental models.
Rigorous validation of EYA1 knockdown is essential for interpreting phenotypic effects. A comprehensive validation strategy includes:
Multiple shRNA constructs:
Multi-level validation:
Specificity controls:
Assess expression of other EYA family members (EYA2-4) to confirm specificity
Test for rescue of phenotypes by expressing shRNA-resistant EYA1 constructs
Compare phenotypes across multiple shRNAs to distinguish on-target from off-target effects
Temporal considerations:
Establish time course of knockdown to correlate with phenotypic changes
Consider inducible knockdown systems for studying developmental processes
Documentation requirements:
Image entire Western blots with molecular weight markers
Quantify knockdown efficiency across biological replicates
Document all experimental conditions in detail for reproducibility
This comprehensive validation approach ensures that observed phenotypes can be confidently attributed to EYA1 depletion rather than off-target effects.
Researchers frequently encounter several challenges when working with EYA1 antibodies:
High background in immunostaining:
Problem: Non-specific staining obscuring true EYA1 signal
Solutions: Increase blocking time/concentration; use alternative blocking agents (BSA, serum, commercial blockers); optimize antibody dilution; include additional washing steps; use monoclonal antibodies for greater specificity
Weak or absent signal in Western blots:
Problem: Inability to detect EYA1 despite expected expression
Solutions: Optimize protein extraction method for nuclear proteins; increase protein loading; reduce transfer time for large proteins; use enhanced detection systems; verify sample preparation preserves EYA1 integrity; try antibodies targeting different epitopes
Multiple bands in Western blots:
Problem: Unclear which band represents genuine EYA1
Solutions: Include positive control samples; perform EYA1 knockdown to identify specific bands; consult literature for expected band patterns; consider post-translational modifications or isoforms
Inconsistent immunoprecipitation results:
Cross-reactivity with other EYA proteins:
Problem: Antibody recognizes multiple EYA family members
Solutions: Verify antibody specificity through knockout/knockdown controls; use epitope-specific antibodies; perform comparative analysis with multiple antibodies; consider computational sequence analysis to identify unique epitopes
Each troubleshooting approach should be systematically documented to establish optimal conditions for your specific experimental system.
Conflicting results from different EYA1 antibodies are not uncommon and require careful interpretation:
Epitope accessibility analysis:
Different antibodies recognize distinct epitopes that may be differentially accessible in various experimental conditions
Map the recognized epitopes to functional domains of EYA1 and consider whether protein interactions or conformational changes might mask specific regions
Test multiple antibodies recognizing different regions in parallel
Cross-reactivity assessment:
Evaluate whether discrepancies result from differential recognition of EYA family members
Perform side-by-side validation in systems with known EYA1 expression versus EYA2-4 expression
Use genetic approaches (siRNA, CRISPR) to validate antibody specificity
Technical validation approaches:
Compare antibody performance across multiple lots and suppliers
Use recombinant EYA1 protein as a positive control
Validate key findings with orthogonal techniques not relying on antibodies (e.g., mass spectrometry)
Contextual interpretation:
Consider that different results may reflect biological reality rather than technical artifacts
EYA1 undergoes post-translational modifications and exists in multiple complexes
Different antibodies may preferentially detect specific EYA1 subpopulations
Resolution strategies:
For critical findings, confirm with at least two independent antibodies
Complement antibody-based methods with genetic approaches
Document all variables that might contribute to differential results
These analytical approaches transform potentially conflicting results into deeper insights about EYA1 biology and methodological considerations.
Robust statistical analysis of EYA1 expression data requires attention to several methodological considerations:
Experimental design fundamentals:
Ensure adequate biological replicates (minimum n=3, preferably n≥5)
Include technical replicates to assess methodological variability
Design experiments to control for batch effects and confounding variables
Normalization strategies:
For Western blot quantification, normalize EYA1 signal to appropriate loading controls (GAPDH, β-actin, total protein)
In immunofluorescence, normalize to nuclear staining or cell area
For qRT-PCR, validate reference genes for stability across experimental conditions
Statistical tests for different experimental scenarios:
Two-group comparisons: t-test (parametric) or Mann-Whitney (non-parametric)
Multiple group comparisons: ANOVA with appropriate post-hoc tests
Correlative studies: Pearson or Spearman correlation coefficients
Time-course experiments: Repeated measures ANOVA or mixed-effects models
Advanced analytical approaches:
For heterogeneous samples (e.g., tumor specimens), consider multiparameter analysis correlating EYA1 with clinical variables
For complex datasets, employ multivariate analysis to identify patterns associated with EYA1 expression
Use machine learning approaches for predictive modeling based on EYA1 and related markers
Reporting requirements:
Clearly state sample sizes, statistical tests, and p-values
Report effect sizes and confidence intervals, not just statistical significance
Include all data points in visualizations, not just means/medians
Document software and analysis parameters for reproducibility
EYA1 has emerged as a promising therapeutic target, particularly in cancer stem cell biology:
Glioblastoma stem cell targeting:
EYA1 is preferentially expressed in glioblastoma stem cells compared to differentiated tumor progeny
Targeting EYA1 decreases GSC proliferation, migration, and self-renewal in vitro and reduces tumor growth in vivo
The EYA1-MYC interaction represents a potential therapeutic vulnerability, as EYA1 targeting leads to loss of MYC expression
Therapeutic development approaches:
Small molecule inhibitors: Targeting EYA1's phosphatase domain
Peptide-based inhibitors: Disrupting protein-protein interactions, particularly with SIX proteins or MYC
Gene therapy approaches: Using shRNA or CRISPR systems for EYA1 inhibition in tumors
Targeting strategies:
Direct targeting of EYA1 activity or expression
Indirect targeting of downstream pathways
Combination approaches targeting both EYA1 and interacting partners like MYC
Biomarker potential:
EYA1 expression as a prognostic or predictive biomarker
EYA1 activity as a measure of therapeutic response
The emerging understanding of EYA1's role in regulating stem-like properties in tumors, particularly through interaction with MYC, provides a compelling rationale for continued investigation as a therapeutic target .
Recent advances have enhanced our ability to study EYA1's developmental functions:
Advanced imaging techniques:
Live imaging using fluorescently-tagged EYA1 to track subcellular localization during development
Super-resolution microscopy to visualize EYA1 interactions with nuclear components
Intravital imaging of EYA1 expression in developing organs in model organisms
Genetic engineering approaches:
CRISPR/Cas9-mediated genome editing to introduce specific mutations in EYA1
Conditional knockout models to study tissue-specific requirements
Creation of reporter lines where fluorescent proteins are expressed under endogenous EYA1 regulatory elements
Single-cell techniques:
Single-cell RNA sequencing to identify EYA1-expressing populations during development
CyTOF and spectral flow cytometry to correlate EYA1 with developmental markers
Spatial transcriptomics to map EYA1 expression in developing tissues
Organoid and 3D culture systems:
Using EYA1 antibodies to study expression and localization in organoids
Assessing effects of EYA1 manipulation on organoid formation and differentiation
Co-culture systems to study cell-cell interactions mediated by EYA1
These methodologies allow researchers to address fundamental questions about EYA1's role in organ development and morphogenesis with unprecedented spatial and temporal resolution.
Recent research has expanded our understanding of EYA1's regulatory networks:
EYA1-MYC regulatory axis:
Integrated pathway analysis:
Cross-talk between EYA1 and major developmental signaling pathways (Notch, Wnt, Sonic Hedgehog)
Integration of growth factor signaling with transcriptional control via EYA1
Combinatorial effects of EYA1 with SIX family members in target gene regulation
Post-translational regulation:
Phosphorylation states affecting EYA1 function
Protein stability regulation through ubiquitin-proteasome system
Subcellular localization dynamics in response to cellular signals
Phosphatase substrate identification:
Novel targets of EYA1's phosphatase activity
Functional consequences of substrate dephosphorylation
Differential substrate preferences in development versus disease contexts
These emerging insights highlight EYA1's position at the intersection of multiple regulatory networks, particularly in stem cell contexts where it may serve as a critical link between external signals and transcriptional responses through mechanisms including its newly identified interaction with MYC .