The PAX8 antibody, conjugated with fluorescein isothiocyanate (FITC), is a widely used research tool for detecting the PAX8 transcription factor in various biological samples. PAX8 is a member of the paired box (PAX) family of transcription factors, critical for organogenesis in the thyroid, kidney, and Mullerian system. This article synthesizes data from diverse sources to provide a detailed analysis of the antibody’s specifications, applications, and research significance.
Host/Isotype: Available as mouse monoclonal (IgG2a Kappa/IgG2b Kappa) or rabbit polyclonal IgG .
Conjugate: FITC (excitation/emission: 495/519 nm) for fluorescence-based assays .
Target: Recognizes a 62 kDa nuclear protein (PAX8) involved in thyroid follicular cell development and tumor pathogenesis .
PAX8 is overexpressed in ovarian serous, endometrioid, and clear cell carcinomas, making it a diagnostic marker for distinguishing ovarian from mammary tumors . The antibody has been used to study PAX8’s interaction with SOX17, which promotes angiogenesis in high-grade serous ovarian cancers (HGSOC) .
PAX8 lineage-driven therapies, such as T-cell engaging bispecific antibodies (TCBs), show promise in targeting LYPD1-expressing HGSOC cells while sparing normal tissues .
PAX8 is a member of the paired box (PAX) family of transcription factors containing a paired box domain and a paired-type homeodomain. It is expressed during organogenesis of the thyroid gland, kidney, and Mullerian system . PAX8's significance in cancer research stems from its role in regulating the expression of Wilms tumor suppressor (WT1) gene, with mutations in PAX8 associated with Wilms tumor cells, thyroid and ovarian carcinomas . It serves as a useful marker in distinguishing ovarian carcinomas from mammary carcinomas, as PAX8 is expressed in a high percentage of ovarian serous, endometrioid, and clear cell carcinomas, but only rarely in primary ovarian mucinous adenocarcinomas . Additionally, PAX8 expression is reported in renal tubules, renal cell carcinoma, nephroblastoma, and seminoma .
FITC-conjugated PAX8 antibodies have specific excitation and emission wavelengths that define their optical properties and applications:
Conjugate | Excitation (nm) | Emission (nm) | Advantages | Limitations |
---|---|---|---|---|
FITC | 495 | 519 | Widely compatible with standard filters, cost-effective | Moderate photobleaching, pH sensitive |
CoraLite 594 | 588 | 604 | Red fluorescence, less tissue autofluorescence | Requires specialized filter sets |
Alexa Fluor 647 | ~650 | ~668 | Far-red spectrum, minimal spectral overlap | Higher cost, requires specific filters |
These spectral characteristics are important when designing multicolor immunofluorescence experiments, as researchers must ensure minimal spectral overlap between fluorophores .
PAX8 antibody, FITC conjugated, is suitable for several research applications with specific recommended dilutions:
For all applications, it is strongly recommended that the antibody be titrated in each specific testing system to obtain optimal results . Sample-dependent variations may occur, necessitating examination of validation data provided by antibody suppliers .
Proper storage of FITC-conjugated PAX8 antibody is crucial for maintaining its fluorescence intensity and binding specificity:
Temperature: Store at -20°C for long-term storage or 4°C in the dark for short-term use
Light protection: Crucially important as FITC is photosensitive; avoid exposure to light during storage and handling
Buffer composition: Typically stored in PBS with additives such as 50% Glycerol, 0.05% Proclin300, 0.5% BSA, pH 7.3
Stability: Generally stable for one year after shipment when stored properly
Aliquoting considerations: While some sources mention aliquoting is unnecessary for -20°C storage , creating small working aliquots is generally recommended practice to avoid repeated freeze-thaw cycles
For FITC-conjugated antibodies specifically, protection from light is particularly important as exposure to light accelerates photobleaching of the fluorophore, which can significantly reduce signal intensity during experiments.
Optimizing immunofluorescence staining with FITC-conjugated PAX8 antibody requires attention to several critical parameters:
Fixation and Permeabilization:
Use 4% formaldehyde fixation for 10 minutes followed by permeabilization with 0.1% Triton X-100 for 5 minutes
For nuclear transcription factors like PAX8, efficient permeabilization is essential for antibody access
Blocking:
Implement 1% BSA/10% normal goat serum/0.3M glycine in 0.1% PBS-Tween for 1 hour to reduce background fluorescence
The blocking solution should match the host species of the secondary antibody if using a detection system
Antibody Dilution and Incubation:
For nuclear antigens like PAX8, overnight incubation at 4°C often yields better signal-to-noise ratios
Include positive controls from known PAX8-expressing cells (SKOV-3 or OVCAR-3 cells are recommended)
Nuclear Counterstaining:
DAPI is commonly used for nuclear counterstaining, which helps verify the nuclear localization of PAX8
The nuclear counterstain should have minimal spectral overlap with FITC
Mounting Considerations:
Use an anti-fade mounting medium specifically formulated to preserve FITC fluorescence
Seal edges of coverslips to prevent drying and oxidation of the fluorophore
For challenging samples or when higher sensitivity is required, consider implementing a signal amplification system, though this should be carefully validated to ensure specificity is maintained.
Proper controls are crucial for generating reliable and interpretable data with FITC-conjugated PAX8 antibodies:
Positive Controls:
Cell lines with known PAX8 expression: SKOV-3, OVCAR-3, or thyroid carcinoma cell lines
Tissue sections from thyroid, kidney, or fallopian tube epithelium
These controls validate the antibody's ability to detect the target protein
Negative Controls:
PAX8-negative cell lines or tissues
Isotype control antibodies (matching the primary antibody's host species and isotype) at equivalent concentration
These controls help establish thresholds for non-specific binding
Technical Controls:
Unstained samples to assess autofluorescence
For flow cytometry: Fluorescence Minus One (FMO) controls to set accurate gates
For multicolor imaging: single-color controls for spectral compensation
Specificity Validation Controls:
Pre-absorption with immunizing peptide when available
Parallel staining with alternative PAX8 antibodies from different clones
Correlation with PAX8 mRNA expression data
siRNA knockdown samples where feasible
Implementing these controls systematically ensures that the observed staining truly represents PAX8 protein rather than artifacts or non-specific binding, which is particularly important in publications and when establishing new protocols.
Designing effective multicolor flow cytometry panels with FITC-conjugated PAX8 antibody requires strategic planning to minimize spectral overlap and maximize information content:
Spectral Considerations:
FITC has excitation maximum at 495 nm and emission at 519 nm
Avoid fluorophores with significant spectral overlap such as PE or GFP
Compatible fluorophores include APC, PE-Cy7, BV421, and PerCP-Cy5.5
Example Panel Design:
Marker | Fluorophore | Cell Compartment | Function |
---|---|---|---|
PAX8 | FITC | Nuclear | Transcription factor |
CD45 | APC | Cell Surface | Hematopoietic marker (negative in epithelial cells) |
Cytokeratin | PE-Cy7 | Cytoplasmic | Epithelial marker |
DAPI | BV421 | Nuclear | Viability/DNA content |
Staining Protocol Considerations:
For nuclear antigens like PAX8, implement a sequential staining approach:
Surface markers first (if applicable)
Fixation (4% formaldehyde, 10 minutes)
Permeabilization (0.1% Triton X-100, 5 minutes)
Nuclear staining with PAX8-FITC
Use the recommended concentration of 0.20 μg per 10^6 cells in 100 μl suspension
Essential Controls:
Include single-color controls for compensation
FMO controls for accurate gating
Isotype controls at identical concentrations
Positive and negative biological controls
Analysis Strategy:
Gate sequentially: viable cells → single cells → target population → PAX8 expression
Consider PAX8 expression intensity (MFI) alongside percentage of positive cells
For heterogeneous samples, correlate PAX8 expression with other markers to identify specific cell populations
This approach enables accurate quantification of PAX8 expression in complex cellular populations and facilitates comparison between experimental conditions.
Recent research has revealed important interactions between PAX8 and SOX17 in ovarian cancer, with significant implications for understanding disease mechanisms:
Physical Interaction Evidence:
PAX8 and SOX17 physically interact as demonstrated by co-immunoprecipitation studies
This interaction is markedly increased in high-grade serous ovarian carcinoma (HGSOC) compared to fallopian tube epithelial (FTE) cells
Size-exclusion chromatography has shown that PAX8 and SOX17 form part of a larger complex of approximately 600 kDa
Subcellular Co-localization:
High-resolution immunofluorescence analyses have demonstrated nuclear co-localization of PAX8 and SOX17 in:
Proximity Ligation Assay Confirmation:
In situ proximity ligation assay (PLA) has confirmed increased interaction between PAX8 and SOX17 in HGSOC cell lines
These protein-protein interactions are localized in the nuclei, consistent with their roles as transcription factors
Research Implications:
FITC-conjugated PAX8 antibodies provide valuable tools for studying these interactions in multicolor imaging experiments
The increased PAX8-SOX17 interaction in cancer cells suggests altered transcriptional regulation that may contribute to carcinogenesis
Understanding these interactions may reveal new therapeutic targets for ovarian cancer treatment
The discovery of this interaction highlights how PAX8 antibodies contribute to mechanistic studies beyond simple protein detection, advancing our understanding of transcriptional regulation in cancer.
Proximity Ligation Assay (PLA) is a powerful technique for visualizing protein-protein interactions in situ. Adapting this method for use with FITC-conjugated PAX8 antibodies requires specific methodological considerations:
Primary Antibody Selection and Preparation:
When using PAX8 antibody, FITC conjugated for PLA:
Ensure the conjugation doesn't interfere with the antibody's epitope binding
Select an unconjugated antibody against the potential interaction partner (e.g., SOX17)
Verify antibodies are from different host species to enable species-specific secondary antibody recognition
PLA Probe Selection:
Choose PLA probes that emit in spectral regions distinct from FITC (495/519 nm)
Red or far-red emitting PLA probes (e.g., 594 nm or 647 nm) provide good spectral separation
Protocol Steps:
Sample Preparation: Fix cells with 4% formaldehyde (10 min) and permeabilize with 0.1% Triton X-100 (5 min)
Blocking: Use Duolink blocking solution or similar to reduce non-specific binding
Primary Antibody Incubation: Apply both PAX8-FITC and partner antibody simultaneously or sequentially
PLA Probe Application: Use PLA probes specific to the host species of each primary antibody
Ligation and Amplification: Follow standard PLA protocol
Counterstaining: DAPI for nuclear visualization
Analysis Considerations:
The FITC signal identifies PAX8-expressing cells/regions
PLA signals (typically appearing as distinct fluorescent dots) indicate specific interaction sites
Quantify the number and intensity of PLA signals within FITC-positive regions
Compare interaction frequencies between experimental conditions (e.g., normal vs. cancer cells)
This approach has been validated in studies examining PAX8-SOX17 interactions in ovarian cancer, where increased interaction frequency was observed in high-grade serous ovarian carcinoma compared to normal fallopian tube epithelial cells .
Understanding PAX8 expression patterns across normal and cancerous tissues is crucial for diagnostic applications and research interpretations:
Expression Pattern Comparison:
Tissue Type | PAX8 Expression | Detection Characteristics |
---|---|---|
Normal thyroid | Positive | Nuclear staining in follicular cells |
Thyroid carcinomas | Positive (often increased) | Maintained or enhanced nuclear expression |
Normal kidney | Positive in renal tubules | Nuclear staining pattern |
Renal cell carcinoma | Positive | Valuable diagnostic marker |
Normal fallopian tube | Positive in secretory cells | Nuclear staining, negative in ciliated cells |
Ovarian surface epithelium | Negative | Important negative reference point |
Ovarian inclusion cysts | Positive | Contrasts with negative surface epithelium |
Ovarian serous carcinoma | Strongly positive | High percentage of cases show positivity |
Ovarian endometrioid carcinoma | Positive | High percentage of cases |
Ovarian clear cell carcinoma | Positive | High percentage of cases |
Ovarian mucinous adenocarcinoma | Rarely positive | Useful for differential diagnosis |
Breast carcinoma | Negative | Critical for differentiating from ovarian carcinoma |
Methodological Implications:
Antibody Titration: Expression levels vary between tissues, requiring careful antibody titration
Signal Intensity Assessment: Cancer tissues may show altered expression intensity requiring appropriate exposure settings
Background Considerations: Some cancerous tissues have higher autofluorescence requiring stringent controls
Multiplexing Strategy: Co-staining with lineage markers helps distinguish tumor cells in heterogeneous samples
Research Applications:
PAX8-FITC antibodies facilitate identification of tumor origin in metastatic lesions
The differential interaction between PAX8 and SOX17 in normal versus cancer cells provides insight into altered transcriptional networks
Flow cytometric analysis with PAX8-FITC enables quantitative assessment of expression changes during malignant transformation
Correlation of PAX8 expression with clinical outcomes may reveal prognostic significance
These differences underscore PAX8's value as both a diagnostic marker and a research tool for understanding fundamental aspects of carcinogenesis.
When encountering staining issues with FITC-conjugated PAX8 antibodies, a systematic troubleshooting approach is essential:
Addressing Weak Staining:
Antibody Concentration Adjustment:
Antigen Retrieval Enhancement:
For FFPE tissues: Optimize heat-induced epitope retrieval with citrate buffer (pH 6.0) or EDTA buffer (pH 9.0)
For cell lines: Test different permeabilization reagents or extend permeabilization time
Incubation Optimization:
Extend primary antibody incubation (overnight at 4°C rather than 1-2 hours at room temperature)
Ensure consistent temperature during incubation periods
Signal Amplification Strategies:
Consider signal amplification systems compatible with FITC detection
Verify any amplification approach maintains specificity
Resolving Nonspecific Staining:
Blocking Protocol Enhancement:
Washing Procedure Optimization:
Implement additional wash steps or extend washing duration
Ensure wash buffer contains appropriate detergent concentration (0.05-0.1% Tween-20)
Autofluorescence Reduction:
Include autofluorescence quenching step (e.g., 0.1% Sudan Black in 70% ethanol)
For tissue sections: Consider specialized autofluorescence quenchers
FITC-Specific Considerations:
Protect from light during all protocol steps to prevent photobleaching
Verify microscope filter sets are optimized for FITC detection
Check pH of buffers (FITC fluorescence is pH-sensitive)
Essential Controls for Problem Identification:
Negative control (cell line known not to express PAX8)
Isotype control at equivalent concentration
Secondary antibody-only control (if using detection systems)
Systematic evaluation of these factors, combined with careful documentation of modifications, should resolve most staining issues with FITC-conjugated PAX8 antibodies and lead to reproducible, specific detection.
Quantitative analysis of PAX8 expression using FITC-conjugated antibodies in high-throughput screening requires standardized approaches and rigorous controls:
Flow Cytometry-Based Quantification:
Sample Preparation Protocol:
Quantification Metrics:
Mean Fluorescence Intensity (MFI) of PAX8-FITC signal
Percentage of cells above threshold (PAX8-positive population)
Molecules of Equivalent Soluble Fluorochrome (MESF) for standardized cross-experimental comparison
High-Content Imaging Approaches:
Automated Microscopy Setup:
Image Analysis Parameters:
Nuclear PAX8-FITC intensity (integrated or mean)
Nuclear area positive for PAX8 signal
Correlation of PAX8 expression with morphological features
Population heterogeneity assessment
Standardization for Screening Applications:
Quality Control Implementation:
Include standard control wells on each plate (positive, negative, dynamic range)
Calculate Z'-factor to assess assay quality and suitability for screening
Monitor day-to-day variability using reference samples
Data Normalization Strategies:
Percent of control normalization relative to positive and negative controls
Z-score normalization within plates to account for plate-to-plate variation
Consider robust statistical methods resistant to outliers
Application to Functional Genomics:
Correlation of PAX8 expression changes with genetic perturbations (siRNA, CRISPR)
Identification of compounds that modulate PAX8 expression or localization
Integration with other cellular readouts (viability, proliferation, differentiation)
This approach enables reproducible quantification of PAX8 expression across large sample sets, facilitating the identification of factors that regulate this transcription factor in normal and disease contexts.
Detecting PAX8 in ovarian cancer stem cells (CSCs) presents unique challenges that require specific methodological considerations:
Cell Identification and Isolation:
Ovarian CSCs are typically identified using markers such as CD133, CD44, ALDH activity, or sphere-forming ability
Consider whether PAX8 expression might differ between CSC and non-CSC populations
Protocol Optimization for CSC Analysis:
Sample Preparation:
Minimize processing steps that might alter stemness characteristics
Consider gentle fixation protocols (2% paraformaldehyde) to preserve epitopes and CSC markers
Optimize permeabilization to maintain both surface stemness markers and nuclear access
Antibody Titration:
CSCs may express different levels of PAX8 compared to bulk tumor cells
Perform careful titration experiments specifically with purified CSC populations
Multimarker Panel Design:
Marker | Function | Suggested Fluorophore Combination |
---|---|---|
PAX8 | Transcription factor | FITC |
CD133 | CSC marker | APC |
CD44 | CSC marker | PE-Cy7 |
ALDH | Functional CSC marker | Detected via Aldefluor (BAAA) |
Viability dye | Dead cell exclusion | Far-red fluorescent dye |
Analysis Considerations:
Flow Cytometry Approach:
Implement hierarchical gating: viable cells → single cells → CSC marker-positive → PAX8 analysis
Assess whether PAX8 expression correlates positively or negatively with stemness markers
Compare PAX8 expression in matched CSC and non-CSC populations
Imaging Analysis:
Use confocal microscopy for precise localization of PAX8 in CSC populations
Implement z-stack acquisition to fully capture nuclear distribution
Consider intensity quantification in identifiable CSC vs. non-CSC populations
Technical Challenges and Solutions:
CSCs often represent a small percentage of the total tumor cell population, requiring efficient enrichment
Autofluorescence can be problematic; include unstained controls and consider autofluorescence reduction methods
PAX8 expression might be heterogeneous within the CSC population, necessitating single-cell analysis approaches
Validation Strategies:
Confirm PAX8 expression in sorted CSC populations using alternative methods (qPCR, western blot)
Correlate PAX8 expression with functional stemness assays (sphere formation, tumor initiation)
Assess PAX8 expression changes during CSC differentiation experiments
These specialized considerations will enable reliable detection and characterization of PAX8 in ovarian cancer stem cells, potentially revealing its role in stemness maintenance and therapeutic resistance.
Recent antibody-based studies have significantly advanced our understanding of PAX8 biology and function in both normal and pathological contexts:
Protein Interaction Networks:
Biochemical affinity-purification methods have identified novel PAX8-interacting proteins in ovarian carcinoma cell lines (OVCAR4, KURAMOCHI, OVSAHO) and immortalized fallopian tube secretory cells
Size-exclusion chromatography has revealed that PAX8 exists in a complex of approximately 600 kDa
Mass spectrometry analysis has identified SOX17 as a significant PAX8-interacting partner
Differential Interactions in Cancer:
Co-immunoprecipitation studies have demonstrated that the level of PAX8-SOX17 complexes is markedly increased in high-grade serous ovarian carcinoma compared to fallopian tube epithelial cells
Proximity ligation assays have confirmed increased PAX8-SOX17 interactions in HGSOC cell lines
These findings suggest altered transcriptional regulation as a component of ovarian carcinogenesis
Subcellular Localization Insights:
High-resolution immunofluorescence analyses have confirmed the nuclear co-localization of PAX8 and SOX17 in both normal and cancer cell lines
The nuclear localization pattern provides important context for understanding PAX8's function as a transcription factor
Functional Implications:
The discovery of these protein interactions suggests PAX8 participates in larger transcriptional complexes
Different interaction patterns between normal and cancer cells may represent potential therapeutic targets
The specific roles of these interactions in regulating gene expression programs are active areas of investigation
These advances demonstrate how antibody-based approaches continue to reveal new aspects of PAX8 biology beyond simple detection, contributing to our understanding of fundamental biological processes and disease mechanisms.
The choice of PAX8 antibody has significant implications for experimental outcomes and data interpretation in research applications:
Clone-Specific Considerations:
Different antibody clones may recognize distinct epitopes of PAX8, potentially affecting detection in contexts where protein interactions mask specific regions
Monoclonal antibodies like PAX8/1491 + PAX8/1492 offer high specificity but might be sensitive to epitope modifications
Polyclonal antibodies may detect multiple epitopes, providing more robust detection but potentially higher background
Conjugation Effects:
FITC conjugation may affect antibody binding characteristics compared to unconjugated versions
Direct conjugation eliminates secondary antibody steps, reducing background but potentially limiting signal amplification options
The physical properties of the fluorophore (size, charge) can impact tissue penetration and nuclear access
Application-Specific Performance:
An antibody performing well in flow cytometry may not be optimal for immunohistochemistry
Fixation methods can differentially affect epitope accessibility for different antibody clones
Sample type (fresh frozen vs. FFPE) may influence antibody selection decisions
Validation Requirements:
Validation Approach | Purpose | Implementation |
---|---|---|
Multiple antibody comparison | Confirm staining pattern | Use antibodies from different clones/manufacturers |
Genetic validation | Confirm specificity | Test in PAX8 knockdown/knockout systems |
Orthogonal methods | Confirm expression | Correlate with mRNA detection methods |
Positive/negative controls | Establish performance | Include known positive and negative samples |
Research Impact Considerations:
Discrepancies in research findings may sometimes be attributed to antibody differences
Reproducibility challenges can arise from inconsistent antibody selection across studies
Comprehensive reporting of antibody details (clone, manufacturer, lot, dilution) is essential for research transparency
Careful selection of the appropriate PAX8 antibody based on the specific research application, combined with rigorous validation, is crucial for generating reliable and reproducible results in both basic research and clinical studies.