FOXA3 Antibody, HRP conjugated refers to a chemically modified antibody designed for direct detection of the transcription factor FOXA3 (Forkhead Box A3) in biological samples. HRP (Horseradish Peroxidase) conjugation enables enzymatic amplification of signals, streamlining applications like ELISA without requiring secondary antibodies. This format enhances sensitivity and reduces background noise, making it ideal for high-throughput assays .
The HRP-conjugated antibody is primarily used in ELISA to quantify FOXA3 levels in human tissues or cell lysates. This method leverages HRP’s enzymatic activity to catalyze colorimetric reactions, enabling precise measurement of FOXA3 expression .
While direct research on the HRP-conjugated antibody is limited, studies using non-conjugated FOXA3 antibodies highlight its importance:
Liver Regeneration: FOXA3 overexpression in HepaRG cells enhances hepatocyte differentiation and glycogen storage, improving liver function in Fah knockout mice .
Atherosclerosis: Hepatic FOXA3 promotes ApoA-I expression, increasing HDL cholesterol and reducing atherosclerotic lesions in ApoE−/− mice .
| Antibody | Conjugate | Applications | Reactivity | Immunogen |
|---|---|---|---|---|
| ABIN7155058 (HRP-conjugated) | HRP | ELISA | Human | AA 220–336 (Human FOXA3) |
| PA1-813 | Unconjugated | Western Blot, ChIP | Human, Mouse, Rat | Synthetic peptide (Mouse FOXA3) |
| PB9805 | Unconjugated | IHC | Human, Mouse, Rat | C-terminal peptide (Human FOXA3) |
FOXA3 (Forkhead box protein A3), also known as Hepatocyte nuclear factor 3-gamma (HNF-3G) or Transcription factor 3G (TCF-3G), is a transcription factor that acts as a 'pioneer' factor. It opens compacted chromatin for other proteins through interactions with nucleosomal core histones, thereby replacing linker histones at target enhancer and/or promoter sites .
FOXA3 serves multiple critical functions:
Activates transcription for several liver genes including AFP, albumin, tyrosine aminotransferase, and PEPCK
Regulates glucose homeostasis by binding to and activating transcription from the G6PC promoter
Binds to the CYP3A4 promoter and activates its transcription in cooperation with CEBPA
Interacts with the CYP3A7 promoter alongside members of the CTF/NF-I family
Participates in neuronal-specific transcription regulation
Recent research has also implicated FOXA3 in the development of hepatoblastoma (HB) through upregulation of AFP and HNF1A/MYC expression, while downregulating ZFHX3 expression .
FOXA3 Antibody, HRP conjugated is primarily optimized for the following applications:
ELISA (Enzyme-Linked Immunosorbent Assay): The HRP conjugation allows direct detection without secondary antibodies, making it suitable for quantitative measurement of FOXA3 expression levels .
While non-HRP conjugated FOXA3 antibodies can be used for additional applications, with appropriate secondary antibody selection:
Western Blot: Detecting FOXA3 protein expression (approximately 40 kDa observed molecular weight) in cell or tissue lysates .
Immunohistochemistry (IHC): Localizing FOXA3 in paraffin-embedded tissue sections, particularly in liver, pancreatic, and colon tissues .
Immunocytochemistry/Immunofluorescence (ICC/IF): Examining subcellular localization of FOXA3 in cultured cells .
When selecting application-specific protocols, researchers should consider the antibody's validation data and optimize conditions for their specific experimental system.
For optimal performance and longevity of FOXA3 Antibody, HRP conjugated:
Storage: Store at -20°C in a non-frost-free freezer
Buffer composition: Typically supplied in 50% Glycerol, 0.01M PBS, pH 7.4 with 0.03% Proclin 300 as preservative
Aliquoting: Upon receipt, prepare small working aliquots to avoid repeated freeze-thaw cycles
Working dilutions: Prepare fresh working dilutions on the day of use
Light sensitivity: As an HRP-conjugated antibody, protect from prolonged exposure to light
Stability: When properly stored, remains stable for at least 12 months from date of receipt
Reconstitution (if lyophilized): Add appropriate volume of distilled water as specified in product documentation
Improper storage or handling can compromise antibody activity and specificity, leading to experimental variability or failure.
FOXA3 has a calculated molecular weight of 37,140 Da, but the observed molecular weight in Western blot applications is approximately 40 kDa . This discrepancy is typical of many transcription factors and may result from:
Post-translational modifications (phosphorylation, acetylation, etc.)
The presence of charged amino acid residues affecting migration
Protein folding effects on electrophoretic mobility
When using FOXA3 Antibody, HRP conjugated for Western blot:
Expect to observe a band at ~40 kDa
Use appropriate molecular weight markers (30-50 kDa range)
Include positive control samples (e.g., HepG2 whole cell lysates)
Consider running gradient gels (5-20% SDS-PAGE) for optimal resolution
Allow sufficient separation time (2-3 hours at 70V/90V for stacking/resolving gel)
For ELISA applications, this molecular weight information helps in assay design and ensuring that the antibody captures the full-length protein rather than degradation products.
Rigorous experimental design requires appropriate controls for accurate interpretation:
Positive tissue/cell controls:
Negative controls:
Knockdown/knockout validation:
Cross-reactivity assessment:
Test in species other than those validated (human, mouse, rat)
Check for cross-reactivity with other FOXA family members (FOXA1, FOXA2)
Implementing these controls helps distinguish specific signal from background and validates antibody performance in your experimental system.
FOXA3's role as a pioneer transcription factor makes it particularly relevant for chromatin structure research. Here are methodological approaches:
Chromatin Immunoprecipitation (ChIP) assays:
Sequential ChIP (Re-ChIP):
Investigate co-occupancy of FOXA3 with other factors (e.g., CEBPA on CYP3A4 promoter)
Requires careful optimization of elution conditions
ChIP-sequencing (ChIP-seq):
Genome-wide mapping of FOXA3 binding sites
Requires high-quality antibodies with minimal background
Data analysis should focus on motif enrichment and correlation with DNase hypersensitivity
ATAC-seq combined with FOXA3 manipulation:
Compare chromatin accessibility before and after FOXA3 knockdown/overexpression
Can reveal genomic regions dependent on FOXA3 for accessibility
Microscopy techniques:
Immunofluorescence co-localization with histone marks
High-resolution imaging to study FOXA3 dynamics during chromatin opening
These approaches can provide insight into how FOXA3 functions as a pioneer factor in opening chromatin structure.
Recent research has implicated FOXA3 in hepatoblastoma development . To investigate this connection:
FOXA3 knockdown experiments:
siRNA approach (si-FOXA3-1, si-FOXA3-2) to reduce expression
Measure effects on cell viability using CCK-8 assay
Assess colony formation capability
Downstream target analysis:
Monitor expression changes in AFP, HNF1A, MYC, and ZFHX3 following FOXA3 knockdown
Western blot and qRT-PCR for protein and mRNA levels, respectively
Immunohistochemistry in tissue samples
Pathway analysis:
Investigate relationship between FOXA3 and AFP/HNF1A/MYC upregulation
Explore mechanisms of ZFHX3 downregulation
Consider chromatin accessibility at these gene loci
Clinical correlation studies:
Compare FOXA3 expression levels between HB tissues and adjacent normal tissues
Correlate expression with clinical parameters and patient outcomes
Consider FOXA3 as a potential biomarker
In vivo models:
Develop xenograft models with FOXA3 manipulation
Use FOXA3 Antibody, HRP conjugated for IHC analysis of tumor sections
These approaches can help elucidate FOXA3's mechanistic contribution to hepatoblastoma development.
Immunohistochemistry optimization requires tissue-specific considerations:
Antigen retrieval methods:
Tissue-specific parameters:
| Tissue Type | Recommended Antibody Concentration | Incubation Time | Antigen Retrieval | Background Reduction |
|---|---|---|---|---|
| Liver | 2 μg/ml | Overnight, 4°C | EDTA, pH 8.0 | 10% goat serum |
| Colon | 2 μg/ml | Overnight, 4°C | EDTA, pH 8.0 | 10% goat serum |
| Pancreas | 2 μg/ml | Overnight, 4°C | EDTA, pH 8.0 | 10% goat serum |
Detection system optimization:
Counterstaining considerations:
Light hematoxylin counterstain to avoid masking nuclear FOXA3 staining
Adjust counterstaining time based on tissue type
Troubleshooting strategies:
For high background: Increase blocking time/concentration or try different blocking agents
For weak signal: Extend primary antibody incubation or adjust concentration
For non-specific binding: Additional washing steps or higher detergent concentration
These optimization steps ensure reliable FOXA3 detection across different tissue contexts.
When studying FOXA3 function through knockdown approaches:
Multi-level validation approach:
mRNA level: qRT-PCR with FOXA3-specific primers
Protein level: Western blot using FOXA3 Antibody
Functional level: Reporter assays for FOXA3 target genes (AFP, G6PC)
Experimental design recommendations:
Quantitative assessment:
Calculate knockdown efficiency as percentage reduction vs. control
Ensure at least 70-80% knockdown for functional studies
Correlate knockdown level with phenotypic changes
Addressing potential confounding factors:
Check for compensatory upregulation of related factors (FOXA1, FOXA2)
Verify specificity using rescue experiments (re-expression of siRNA-resistant FOXA3)
Consider off-target effects through transcriptome analysis
Integration with functional assays:
This comprehensive validation ensures that observed phenotypes are truly attributable to FOXA3 knockdown.
FOXA3 cooperates with various co-factors to regulate gene expression:
Co-immunoprecipitation (Co-IP) techniques:
Proximity ligation assay (PLA):
In situ detection of FOXA3 interactions with suspected partners
Provides spatial information about interaction in cellular context
Requires specific antibodies for both FOXA3 and its partner(s)
Reporter gene assays:
Construct reporters containing FOXA3 binding sites (e.g., G6PC, CYP3A4 promoters)
Co-transfect with FOXA3 and potential co-factors
Measure additive or synergistic effects on transcriptional activity
DNA-protein complex analysis:
Electrophoretic mobility shift assay (EMSA) with purified factors
Supershift assays using FOXA3 Antibody
DNA pulldown followed by Western blot for co-binding factors
Chromatin studies:
Sequential ChIP to identify co-occupancy at genomic loci
Manipulation of co-factor levels to assess impact on FOXA3 binding
These approaches provide a comprehensive view of FOXA3's cooperative interactions in transcriptional regulation.
Researchers may encounter several technical issues:
High background in immunoassays:
Weak or absent signal:
Multiple bands in Western blot:
Cause: Degradation products, isoforms, or cross-reactivity
Verification: Compare with literature-reported patterns
Solution: Use freshly prepared samples with protease inhibitors
Validation: Confirm specificity with knockdown samples
Reduced HRP activity:
Cause: Repeated freeze-thaw cycles or improper storage
Prevention: Prepare small working aliquots
Testing: Verify HRP activity with substrate-only control
Alternative: Consider using fresh antibody for critical experiments
Signal variability between experiments:
Cause: Inconsistent technique or reagent degradation
Solution: Standardize protocols and reagent preparation
Control: Include internal standards for normalization
Recommendation: Prepare master mixes where possible
Addressing these challenges systematically improves reproducibility and data quality.
Differences between mRNA and protein levels are common for transcription factors and require careful interpretation:
Biological explanations:
Post-transcriptional regulation (miRNAs, RNA-binding proteins)
Post-translational modifications affecting protein stability
Feedback mechanisms regulating FOXA3 levels
Different half-lives of mRNA versus protein
Technical considerations:
Antibody specificity and sensitivity
RNA quality and reverse transcription efficiency
Different dynamic ranges of detection methods
Subcellular localization affecting protein detection
Validation approaches:
Time-course analysis to detect temporal relationships
Inhibitor studies (proteasome, translation inhibitors)
Alternative detection methods (mass spectrometry)
Single-cell analysis to assess population heterogeneity
Data integration framework:
| Observation | Possible Explanation | Validation Approach |
|---|---|---|
| High mRNA, low protein | Rapid protein degradation | Proteasome inhibition |
| Low mRNA, high protein | Protein stability, sensitivity differences | Pulse-chase labeling |
| Changing ratios across conditions | Condition-specific regulation | Time course analysis |
| Tissue-specific discrepancies | Tissue-specific post-transcriptional control | Multi-tissue comparison |
Research context:
Acknowledging these complexities enables more accurate interpretation of experimental data involving FOXA3 expression.
FOXA3's role in glucose homeostasis makes it relevant for metabolic disease research:
Tissue-specific expression analysis:
Compare FOXA3 levels in normal versus diabetic liver tissue
Correlation with glucose-regulating genes (G6PC1, PEPCK)
Relationship with insulin signaling pathway components
Glucose regulation studies:
Nutritional intervention models:
FOXA3 expression changes during fasting/feeding cycles
Response to high-fat diet or ketogenic diet
Integration with other metabolic transcription factors
Therapeutic target assessment:
Small molecule screening for FOXA3 activity modulation
Analysis of FOXA3 binding site polymorphisms in metabolic disease cohorts
Correlation of FOXA3 expression with response to anti-diabetic treatments
Multi-omics integration:
Correlation of FOXA3 chromatin binding with metabolomic profiles
Proteomic analysis of FOXA3 interactome in metabolic disease states
Integration with genome-wide association studies (GWAS) data
These approaches can reveal FOXA3's contributions to metabolic disease pathogenesis and potential therapeutic relevance.
While FOXA3's role in hepatoblastoma is established , its function in other cancers is emerging:
Cancer-type specific expression profiling:
Use FOXA3 Antibody, HRP conjugated for tissue microarray analysis
Compare expression across cancer types and stages
Correlate with patient outcomes and treatment response
Mechanistic studies:
Epigenetic landscape analysis:
FOXA3 binding at cancer-relevant gene promoters
Correlation with DNA methylation patterns
Histone modification changes at FOXA3 binding sites
Drug response modulation:
FOXA3 expression changes following chemotherapy/targeted therapy
Impact of FOXA3 knockdown on drug sensitivity
Combination approaches targeting FOXA3 networks
Emerging technologies:
Single-cell analysis of FOXA3 expression heterogeneity within tumors
Spatial transcriptomics to map FOXA3 expression in tumor microenvironment
CRISPR screens to identify synthetic lethal interactions with FOXA3
These methodologies expand our understanding of FOXA3's roles beyond liver cancer to other malignancies where pioneer transcription factors contribute to disease progression.