EPHA8 belongs to the Eph receptor family, which regulates cell-cell interactions during nervous system development. It binds ephrin-A ligands (A2, A3, A5) and facilitates short-range axonal guidance . Notably, EPHA8 enhances integrin-mediated adhesion to fibronectin through phosphatidylinositol 3-kinase (PI3K) activation, independent of its tyrosine kinase activity . This mechanism involves membrane localization of p110γ PI3K, enabling lipid substrate access for downstream signaling .
The Biotin-conjugated EPHA8 antibody enables:
High-Throughput Screening: Biotin-streptavidin systems amplify detection signals in ELISA, ideal for low-abundance EPHA8 quantification .
Western Blot Analysis: Identifies EPHA8 (~110 kDa) in human and mouse lysates .
Mechanistic Studies: Used to investigate EPHA8’s kinase-independent regulation of integrins and PI3K pathways .
| Feature | EPHA8 Biotin Conjugated | Unconjugated EPHA8 Antibodies |
|---|---|---|
| Detection Sensitivity | Enhanced via streptavidin-HRP | Requires secondary antibodies |
| Multiplexing Potential | High (compatible with other biotinylated probes) | Limited |
| Typical Use Cases | High-throughput assays, co-staining | General immunohistochemistry |
Integrin Activation: EphA8 expression in NIH 3T3 cells increases adhesion to fibronectin via α5β1 and β3 integrins, a process blocked by PI3K inhibitors like wortmannin .
Kinase-Independent Signaling: Both wild-type and kinase-inactive EphA8 mutants promote cell adhesion, underscoring a non-catalytic role for EPHA8 in cytoskeletal regulation .
PI3K Association: EphA8 binds p110γ PI3K via its juxtamembrane domain, facilitating membrane localization and lipid phosphorylation .
EPHA8 (Ephrin type-A receptor 8) is a receptor tyrosine kinase belonging to the Eph family, which constitutes the largest family of tyrosine kinase receptors in the human genome. EPHA8 functions as a receptor that binds promiscuously to GPI-anchored ephrin-A family ligands (particularly EFNA2, EFNA3, and EFNA5) on adjacent cells, leading to contact-dependent bidirectional signaling .
Its significance stems from its roles in:
Regulating integrin-mediated cell adhesion and migration on fibronectin substrate
Promoting neurite outgrowth during neural development
Axon guidance in the developing nervous system
Participating in cell-cell communication via both forward signaling (downstream of the receptor) and reverse signaling (downstream of the ephrin ligand)
Notably, EPHA8 can function through both kinase-dependent and kinase-independent mechanisms, making it an intriguing target for understanding diverse cellular signaling pathways .
Biotin conjugation to EPHA8 antibodies introduces measurable effects on antibody functionality that researchers must account for in experimental design:
When working with biotin-conjugated EPHA8 antibodies, researchers should validate each lot by comparing binding activity at different conjugation ratios to optimize experimental conditions.
Verifying antibody specificity is crucial for experimental validity. For EPHA8 antibodies, employ these complementary approaches:
Cross-reactivity assessment against other Eph family members:
Flow cytometry validation:
Knockout/knockdown controls:
Use CRISPR/Cas9-mediated EPHA8 knockout cells or siRNA knockdown models
Compare antibody binding between wild-type and knockout/knockdown samples
Immunoprecipitation followed by mass spectrometry:
Competitive binding assays:
Pre-incubate with free recombinant EPHA8 to block specific binding sites
A specific antibody will show significantly reduced signal following this competition
Immunoblotting for size verification:
For successful co-immunoprecipitation (Co-IP) experiments with biotin-conjugated EPHA8 antibodies:
Sample preparation optimization:
Lyse cells in non-denaturing buffers (e.g., RIPA or NP-40 buffer with protease/phosphatase inhibitors)
For EPHA8 membrane protein complexes, include 1% digitonin or 0.5% CHAPS to preserve protein-protein interactions
Strategic conjugation approach:
Capture method selection:
Detection strategy:
Avoid using biotin-based detection methods that would interfere with the conjugated antibody
Use HRP-conjugated antibodies against expected interacting partners (e.g., anti-p110γ PI 3-kinase)
Consider using a non-biotin conjugated EPHA8 antibody recognizing a different epitope for verification
Controls implementation:
Validation approach:
Confirm co-precipitation using reciprocal IP (i.e., IP with partner protein antibody, detect EPHA8)
Analyze co-IP samples by western blot probing for known EPHA8 interacting proteins
Distinguishing kinase-dependent from kinase-independent EPHA8 functions requires careful experimental design:
Kinase-inactive EPHA8 mutant expression:
Phosphorylation status analysis:
Use phospho-specific antibodies against EPHA8 tyrosine residues
Perform western blotting to verify phosphorylation state differences between wild-type and kinase-inactive forms
For precise quantification, combine with phosphopeptide mass spectrometry
Domain-specific mutants utilization:
Integrin-mediated adhesion assays:
PI 3-kinase signaling pathway investigation:
Ephrin ligand stimulation:
EPHA8 regulation of integrin activity through p110γ PI 3-kinase represents a key kinase-independent pathway that can be investigated through several approaches:
Biochemical interaction characterization:
Functional analysis protocol:
PI 3-kinase inhibition studies:
Dominant-negative approach:
Ligand stimulation experiments:
Integrin activation measurement:
Use antibodies recognizing activated integrin conformations (e.g., HUTS-21 for activated β1)
Perform flow cytometry to quantify integrin activation levels
Compare between controls and cells expressing EPHA8 variants
Proper controls are critical for immunofluorescence studies using biotin-conjugated EPHA8 antibodies:
Specificity controls:
Negative control tissues/cells known to lack EPHA8 expression
Competitive inhibition using recombinant EPHA8 protein to block specific binding
Secondary antibody-only control to assess background
Signal validation approaches:
Compare staining pattern with a second non-biotin conjugated EPHA8 antibody targeting a different epitope
Use EPHA8 knockout or knockdown samples as negative controls
Validate subcellular localization by co-staining with established markers for relevant cellular compartments
Biotin-specific considerations:
Block endogenous biotin using avidin/biotin blocking kits before applying biotin-conjugated antibodies
Include controls for potential streptavidin binding to endogenous biotinylated proteins
Use titratable detection approaches to optimize streptavidin-fluorophore concentration
Technical controls:
Use an isotype-matched biotin-conjugated irrelevant antibody as negative control
Include positive controls with known EPHA8 expression patterns
Test for streptavidin detection system non-specific binding
Cross-reactivity assessment:
Stain tissues from EPHA8 knockout mice to confirm absence of cross-reactivity with other Eph family members
Test antibody on cells expressing individual EphA family members to ensure specificity
To systematically assess the impact of biotin conjugation load on EPHA8 antibody properties:
Preparation of defined conjugation series:
Thermostability measurement:
Binding activity quantification:
Functional activity assessment:
Storage stability analysis:
Fc receptor binding assessment:
Investigating EPHA8's interaction with other Eph receptors requires specialized techniques:
Co-immunoprecipitation with differential tagging:
Proximity ligation assay (PLA):
Use specific antibodies against EPHA8 and potential partner Eph receptors
PLA signal indicates close proximity (<40 nm) suggesting heterooligomerization
Quantify PLA dots per cell to measure interaction frequency
Receptor biotinylation and selective recovery:
Engineer Eph receptors containing N-terminal biotin acceptor peptide (AP) tags
Express with E. coli biotin ligase (BirA) for selective biotinylation
Use streptavidin-coated magnetic Dynabeads to cluster and recover complexes
Western blot with α-EPHA8 antibodies to detect association with other biotinylated Eph receptors
Förster resonance energy transfer (FRET):
Create fusion proteins of EPHA8-CFP and other Eph receptors-YFP
Measure FRET efficiency to quantify protein-protein interactions
Live-cell imaging allows visualization of interactions in real-time
Bimolecular fluorescence complementation (BiFC):
Fuse EPHA8 with one half of a fluorescent protein (e.g., YFP-N)
Fuse other Eph receptors with complementary half (e.g., YFP-C)
Fluorescence occurs only when proteins interact to reconstitute the fluorophore
Size exclusion chromatography coupled with multi-angle light scattering (SEC-MALS):
Analyze purified receptor ectodomains to determine oligomeric states
Compare size distributions of individual receptors versus mixtures
Shifts in molecular weight indicate complex formation
Single-molecule imaging techniques:
Use total internal reflection fluorescence (TIRF) microscopy with differentially labeled receptors
Track co-diffusion and co-localization at the single-molecule level
Quantify interaction dynamics and duration
For studying EPHA8's role in axonal development:
Developmental timing analysis:
Reporter gene approaches:
Axon tracing combined with immunostaining:
Apply anterograde tracers (e.g., DiI) to axonal origins
Co-stain with EPHA8 antibodies to correlate expression with specific projections
Compare patterns between wild-type and EPHA8-mutant mice
Explant culture systems:
Culture tissue explants from regions with EPHA8 expression
Treat with EPHA8-blocking antibodies or ephrin-A ligands
Measure effects on axonal outgrowth direction and extent
Functional perturbation approaches:
Time-lapse imaging with combined labeling:
Culture neurons expressing fluorescent proteins
Apply labeled EPHA8 antibodies to track receptor dynamics
Monitor growth cone behavior in response to ephrin gradients
When investigating EPHA8 in tumor contexts:
Multiplex immunofluorescence protocol optimization:
Signal amplification strategies:
Utilize tyramide signal amplification for low-abundance targets
Compare direct detection versus amplified detection methods
Optimize streptavidin-fluorophore concentration and incubation time
Tissue sample preparation considerations:
Fresh frozen versus formalin-fixed paraffin-embedded tissues require different protocols
For FFPE samples, optimize antigen retrieval methods (citrate buffer pH 6.0 versus EDTA pH 9.0)
Include proper controls for tissue autofluorescence
Quantitative image analysis approaches:
Flow cytometry panel design:
Experimental validation in immunodeficient models:
When facing reproducibility challenges:
Antibody validation protocol implementation:
Verify epitope specificity using peptide competition assays
Compare results across multiple antibody lots and clones
Test on positive and negative control samples with known EPHA8 expression status
Biotin conjugation quality assessment:
Storage condition optimization:
Buffer compatibility testing:
Some buffers may interfere with biotin-streptavidin interaction
Test antibody performance in different buffer systems
Optimize buffer components (salt concentration, pH, detergents)
Detection system troubleshooting:
Compare different streptavidin conjugates (HRP, fluorophores)
Check for streptavidin binding to endogenous biotinylated proteins
Use avidin/biotin blocking kits to reduce background
Systematic protocol optimization:
Titrate antibody concentration to determine optimal working dilution
Try different incubation conditions (time, temperature)
For western blotting, test various blocking agents (BSA vs. milk proteins)
For ELISA, recommended dilutions are typically 1:10,000, while for western blotting, 1:500 may be more appropriate