The AVIL antibody is a highly specific immunological tool designed to detect and study the Advillin protein (encoded by the AVIL gene), a member of the gelsolin/villin family of actin-binding proteins. These proteins regulate actin cytoskeleton dynamics, which are critical for cellular processes such as migration, proliferation, and morphogenesis. AVIL is primarily expressed in neuronal cells and sensory neurons, with emerging roles in pathological conditions like glioblastoma .
Protein Structure: AVIL contains a conserved villin-like headpiece domain responsible for actin binding, enabling it to modulate actin filament assembly and disassembly .
Biological Roles: AVIL regulates neuronal development, particularly in ganglion formation, and has been implicated in tumorigenesis through FOXM1 stabilization .
Glioblastoma Studies: AVIL overexpression correlates with poor patient survival (median 23.1 months vs. 75.1 months in low-expression groups, p < 10^-5) . Antibodies like Abcam’s Ab72210 (used in immunohistochemistry) validate these findings .
Neuronal Development: AVIL antibodies (e.g., Boster Bio A05371) enable visualization of actin-binding dynamics via immunofluorescence, revealing cytoskeletal remodeling in sensory neurons .
Survival Prognosis: High AVIL expression in glioblastoma tissues predicts aggressive tumor behavior, making it a candidate biomarker for prognosis and therapeutic targeting .
Therapeutic Implications: Silencing AVIL via siRNA or shRNA inhibits glioblastoma cell proliferation and migration, suggesting potential for targeted therapies .
AVIL (Advillin) is a member of the gelsolin/villin family of actin regulatory proteins with significant structural similarity to villin. It functions primarily in controlling actin filament assembly, cell migration, and cell adhesion, making it a critical player in fundamental cellular processes. Research has revealed that AVIL is frequently overexpressed in cancer cells, particularly in glioblastoma (GBM) and rhabdomyosarcoma (RMS), where it drives tumorigenesis through multiple mechanisms . AVIL binds to actin and may play important roles in the development of neuronal cells that form ganglia . Its significance lies in both understanding fundamental cellular processes and as a potential therapeutic target in oncology.
AVIL antibodies have been validated for multiple experimental applications with specific methodological considerations for each:
| Application | Typical Dilutions | Sample Preparation Notes | Common Detectable Forms |
|---|---|---|---|
| Western Blot (WB) | 1:500-1:2000 | Standard protein extraction | ~92-111 kDa band (may vary) |
| Immunohistochemistry (IHC) | 1:100-1:200 | TRIS-EDTA-boric acid (pH 8.4) retrieval | Cellular expression patterns |
| Immunofluorescence (IF/ICC) | 1:50-1:200 | 4% PFA fixation recommended | Subcellular localization |
| ELISA | 1:20000-1:80000 | Varies by kit | Quantitative detection |
For optimal results, researchers should consider antigen retrieval methods like heat-induced epitope retrieval using TRIS-EDTA-boric acid buffer (pH 8.4) for IHC applications, particularly when working with formalin-fixed, paraffin-embedded (FFPE) specimens .
AVIL antibodies require specific storage and handling protocols to maintain their reactivity and specificity:
Long-term storage: Maintain at -20°C in small aliquots to prevent repeated freeze-thaw cycles, which can degrade antibody quality
Short-term use: Store at 4°C for up to one month for frequent usage
Reconstitution: For lyophilized antibodies, reconstitute in 100 μl of sterile distilled H₂O with 50% glycerol
Working solution preparation: When diluting for experiments, use fresh, high-quality buffers
Contamination prevention: Work with aseptic techniques to prevent microbial growth
Research indicates that antibody stability decreases significantly after 3-5 freeze-thaw cycles, so preparing single-use aliquots upon receipt is strongly recommended for maintaining consistent experimental results .
For robust AVIL detection in glioblastoma samples, researchers should implement a multi-faceted approach:
Immunohistochemistry protocol optimization:
Deparaffinize tissue sections using a validated protocol (e.g., EZ Prep solution)
Perform heat-induced antigen retrieval for 64 minutes using TRIS-EDTA-boric acid pH 8.4 buffer
Block endogenous peroxidases (e.g., with CM1 for 8 minutes)
Incubate with AVIL antibody at 1:100 dilution for 60 minutes at room temperature
Use anti-rabbit HQ HRP detection system and appropriate chromogen (e.g., DAB)
Scoring system for AVIL expression:
Implement a semiquantitative analysis on a scale of 0-3:
Complementary validation approaches:
These protocols have been successfully implemented in research demonstrating the clinical significance of AVIL in glioblastoma, where AVIL protein expression showed a strong inverse correlation with patient survival (R = −0.82, p = 0.0012) .
Optimizing Western blot conditions for AVIL detection requires careful attention to several parameters:
Sample preparation:
Gel selection and separation:
Transfer conditions:
For large proteins like AVIL, implement longer transfer times or utilize semi-dry transfer systems
Verify transfer efficiency with reversible staining before blocking
Antibody conditions:
Detection method:
Use enhanced chemiluminescence with exposure optimization
For quantitative analysis, consider fluorescence-based Western detection systems
Researchers should expect to observe AVIL at approximately 92-111 kDa, though the observed molecular weight may vary slightly between tissue types and experimental conditions .
Rigorous validation of AVIL antibody specificity requires comprehensive controls:
Positive tissue/cell controls:
Negative tissue/cell controls:
Molecular validation controls:
Technical controls:
Research demonstrates that effective AVIL antibody validation should show absence of signal after AVIL knockdown and increased signal with overexpression, confirming specificity. For example, studies have validated AVIL antibodies using knockdown approaches that showed complete absence of signal following efficient silencing .
AVIL expression demonstrates significant correlations with clinical outcomes in glioblastoma patients across multiple parameters:
Survival correlation:
Strong inverse correlation between AVIL protein expression and patient survival (R = −0.82, p = 0.0012)
Kaplan-Meier analyses show poor prognosis associated with high AVIL protein expression (log-rank test p = 0.0005)
In REMBRANDT project data (343 glioma cases), elevated AVIL expression correlated with shorter survival (upregulated vs. intermediate, p = 1 × 10⁻⁵; upregulated vs. all other, p = 4 × 10⁻⁷, log-rank test)
Prognostic stratification:
Histopathologic correlation:
These findings suggest that AVIL expression assessment using validated antibodies may serve as a valuable prognostic biomarker in clinical neuro-oncology, potentially outperforming conventional histopathological approaches .
AVIL plays crucial roles in tumor cell migration and invasion through multiple mechanistic pathways:
Cytoskeletal regulation:
Experimental evidence of migration dependency:
Wound-healing assays showed dramatic reduction in cell movement when AVIL was silenced in GBM cell lines (A172 and U251)
Transwell invasion assays demonstrated significantly reduced invasiveness following AVIL knockdown
Live-cell imaging tracking individual cells confirmed reduced motility with AVIL silencing
Metastatic potential:
Molecular interactions:
These findings suggest that antibody-based detection of AVIL expression may help predict invasive potential of tumors, providing valuable prognostic information beyond simple proliferation markers .
AVIL antibodies serve as critical tools for investigating oncogene addiction mechanisms through multiple experimental approaches:
Cell survival dependency studies:
AVIL antibodies can quantify expression levels before and after knockdown experiments
Research shows GBM cells die when AVIL is silenced, while normal astrocytes (low AVIL) remain unaffected
This differential effect demonstrates classical oncogene addiction, where cancer cells become dependent on AVIL overexpression
Xenograft model analyses:
AVIL antibodies enable immunohistochemical assessment of tumor formation in animal models
Studies using U251 intracranial xenograft models showed that silencing AVIL prevented tumor formation
Control animals developed significant tumor volumes within 4 weeks, while shAVIL animals showed minimal or no tumor formation by MRI
Transformation capacity assessment:
Antibodies help monitor AVIL levels during transformation experiments
AVIL overexpression induced focus formation in NIH3T3 cells at greater rates than established oncogenic factors (EGFR vIII mutant, shRNA targeting TP53, or shRNA targeting RB)
Astrocytes overexpressing AVIL formed colonies in soft agar assays and developed tumors when injected into nude mice, confirming transformation
Pathway interaction studies:
These methodologies underscore how AVIL antibodies enable comprehensive investigation of oncogene addiction mechanisms, potentially guiding development of targeted therapies for cancers dependent on AVIL overexpression .
Different AVIL antibodies demonstrate variable epitope recognition and performance characteristics that researchers should consider when selecting reagents:
Performance considerations:
Epitope accessibility: Antibodies targeting different regions may perform differently depending on protein folding and complex formation
Cross-reactivity: Antibodies against highly conserved domains may cross-react with related proteins in the gelsolin/villin family
Application-specific performance: Some antibodies excel in certain applications but perform poorly in others
Researchers in the field have found that antibodies recognizing the C-terminal region (including the headpiece domain) are particularly valuable for functional studies, as this region mediates critical actin-binding interactions required for AVIL's oncogenic properties .
Developing a robust immunohistochemical scoring system for AVIL in tumor samples requires attention to several critical factors:
Standardized staining protocol:
Scoring system design:
Quality control measures:
Include known positive and negative controls with each staining batch
Implement blinded scoring by multiple pathologists to ensure reproducibility
Calculate inter-observer concordance statistics
Document representative images for each score level
Clinical correlation methodology:
Correlate scores with patient survival data using Kaplan-Meier analyses
Calculate hazard ratios through multivariate analyses
Determine optimal cutoff values using receiver operating characteristic curves
Assess prognostic value independent of established markers
Research has validated that AVIL immunohistochemical scoring following these principles provides significant prognostic information. For example, high AVIL protein expression determined through such scoring systems demonstrated significant correlation with poor prognosis (log-rank test p = 0.0005) .
Integrating AVIL mutation studies with antibody-based detection requires sophisticated methodological approaches:
Epitope-specific antibody selection:
Choose antibodies that recognize regions away from common mutation sites
Consider using multiple antibodies recognizing different domains
For known mutations, develop mutation-specific antibodies when feasible
Functional domain analysis:
Combined genomic and proteomic approaches:
Sequence AVIL gene from tumor samples to identify mutations
Correlate mutation status with protein expression patterns detected by antibodies
Perform immunoprecipitation followed by mass spectrometry to identify post-translational modifications
Experimental validation protocols:
Generate expression vectors containing wild-type and mutant AVIL
Perform rescue experiments in AVIL-silenced cells
Compare antibody detection patterns between wild-type and mutant proteins
Assess functional outcomes (proliferation, migration) for correlation with antibody signals
Research demonstrates that mutant AVIL proteins (K808C and F819C) with reduced actin binding fail to promote cell proliferation and migration at rates comparable to wild-type AVIL, highlighting how mutation studies can be integrated with functional and antibody-based analyses .
Researchers frequently encounter specific challenges when working with AVIL antibodies:
Nonspecific binding issues:
Variable detection of isoforms:
Fixation-sensitive epitopes:
Cross-reactivity with related proteins:
Problem: Signal in tissues known to lack AVIL expression
Solution: Validate antibody specificity with peptide competition assays
Alternative: Confirm results with orthogonal methods (qRT-PCR, mass spectrometry)
Batch-to-batch variability in polyclonal antibodies:
Problem: Inconsistent results between antibody lots
Solution: Purchase larger quantities of a single lot for long-term studies
Recommendation: Validate each new lot against previous standards and controls
Research demonstrates the importance of thorough validation, as exemplified in studies where AVIL antibody specificity was confirmed through multiple approaches, including siRNA knockdown experiments showing complete absence of signal following efficient silencing .
Interpreting discrepancies between AVIL mRNA and protein expression requires methodological rigor and consideration of multiple biological factors:
These considerations are essential for accurate interpretation, as demonstrated in research where AVIL protein expression proved more clinically relevant than mRNA expression for predicting patient outcomes in glioblastoma .
A comprehensive workflow for validating new AVIL antibodies in cancer research should follow these sequential steps:
Initial characterization:
Verify immunogen sequence conservation across species of interest
Check for potential cross-reactivity with related proteins in silico
Determine optimal antibody concentration through titration experiments
Assess performance across applications (Western blot, IHC, IF, IP) using standard protocols
Specificity validation:
Perform Western blot on cell lines with known AVIL expression (GBM cells as positive controls, normal astrocytes as negative controls)
Conduct knockdown experiments using established siRNAs (siAVIL1 targeting 5′-GCTTCTGGCAAAGGATATT-3′ or siAVIL2 targeting 5′-GCATTCCTTGCTTGTTATA-3′)
Compare with existing validated antibodies if available
Perform peptide competition assays to confirm epitope specificity
Application-specific optimization:
Functional validation:
Correlate antibody signal with known biological functions of AVIL
Test ability to detect changes in expression following experimental manipulation
Verify correlation with clinical parameters in patient samples
Assess reproducibility across multiple experiments and users
This rigorous validation workflow ensures reliable antibody performance in challenging research applications and has been successfully employed in studies investigating AVIL's role in glioblastoma and rhabdomyosarcoma tumorigenesis .
AVIL antibodies offer significant potential in the development of targeted cancer therapies through multiple research pathways:
Target validation studies:
AVIL antibodies can quantify expression across tumor types to identify high-expressing cancers
Immunohistochemical screening of patient-derived xenografts can prioritize cancer types for therapeutic development
AVIL dependency studies show cancer cells are "addicted" to AVIL overexpression, making it an ideal therapeutic target
Therapeutic antibody development:
Investigate intracellular delivery mechanisms for AVIL-targeting antibodies
Explore antibody-drug conjugate approaches targeting AVIL-expressing cells
Develop conformation-specific antibodies that disrupt AVIL-actin interactions
Functional domain targeting:
Biomarker applications:
Implement AVIL antibody-based companion diagnostics to identify patients likely to respond to AVIL-targeted therapies
Develop standardized immunohistochemical protocols for patient stratification
Monitor treatment response through serial assessment of AVIL expression
These approaches are supported by research demonstrating that silencing AVIL induced GBM cell death in vitro and prevented/reduced GBM xenograft formation in animal models, while normal astrocytes remained unaffected—suggesting a high therapeutic index for AVIL-targeted therapies .
Cutting-edge techniques for investigating AVIL-partner interactions include:
Advanced co-immunoprecipitation approaches:
Live-cell interaction visualization:
Structural biology approaches:
Functional genomics integration:
These methodologies offer deeper insights into AVIL's molecular functions, as demonstrated in research where AVIL was shown to potentially regulate FOXM1 stability, linking it to broader oncogenic signaling networks .
Standardizing AVIL detection methods requires collaborative approaches to establish consensus protocols:
Antibody standardization initiatives:
Establish reference standards for commercially available AVIL antibodies
Conduct multi-laboratory validation studies to assess reproducibility
Create shared repositories of validated protocols with detailed methodological parameters
Quantitative assessment frameworks:
Develop digital pathology approaches for objective AVIL quantification
Establish standardized immunohistochemical scoring systems :
0 = 0% cells positive
1 = >0% <10% cells positive
2 = >10% <50% cells positive
3 = >50% cells positive
Implement automated image analysis algorithms for consistent evaluation
Quality control program implementation:
Create proficiency testing programs for AVIL detection
Develop reference materials with known AVIL expression levels
Establish minimum validation requirements for publication-quality data
Harmonized reporting standards:
Adopt standardized nomenclature for AVIL detection methods
Implement detailed reporting of antibody information (source, catalog number, lot, dilution)
Report all validation measures undertaken for novel applications