PAX8 (Paired Box 8) is a nuclear transcription factor belonging to the PAX family, essential for thyroid follicular cell development and renal tubule differentiation . The HRP-conjugated PAX8 antibody binds specifically to PAX8 epitopes, facilitating visualization via chromogenic or chemiluminescent substrates. Key features include:
Conjugate: Horseradish peroxidase (HRP) for signal amplification .
Species Reactivity: Human, mouse, rat, dog, and others, depending on the clone .
PAX8 Antibody, HRP conjugated, is widely used in:
Renal Cell Carcinoma (RCC): Detects PAX8 in 80% of clear cell RCCs, 95% of papillary RCCs, and 100% of chromophobe RCCs .
Thyroid Carcinomas: Identifies thyroid-specific gene expression in follicular and anaplastic thyroid cancers .
Ovarian Cancers: Distinguishes non-mucinous ovarian carcinomas (e.g., serous, endometrioid) from mucinous subtypes .
Quantifies PAX8 in nuclear extracts and co-immunoprecipitates with SOX17 in ovarian cancer studies .
IHC-P: Antigen retrieval with Tris/EDTA (pH 9.0) , blocking with 3% H₂O₂-methanol , and detection via DAB chromogen .
WB: Dilution ranges from 1:500 to 1:10,000 , using HRP-conjugated secondary antibodies (e.g., GTX213110-01) .
A cohort study (n=272 tumors) demonstrated:
PAX8 physically interacts with SOX17 in high-grade serous ovarian carcinoma (HGSOC), promoting VEGF-A secretion and endothelial cell proliferation .
False Negatives: Clear cell RCC shows lower PAX8 expression than papillary/chromophobe subtypes .
Cross-Reactivity: Some clones exhibit non-specific binding at ~35 kDa in WB .
Storage: HRP conjugates require light-protected storage at 4°C to preserve activity .
PAX8 is a member of the paired box (PAX) family of transcription factors with a molecular weight of approximately 62 kDa. It functions as a transcription factor for thyroid-specific gene expression and maintains the functional differentiation of thyroid cells . PAX8 is also expressed in non-ciliated mucosal cells of fallopian tubes, renal tubules, and various cancers including ovarian, renal cell carcinoma, and nephroblastoma .
Its importance in research stems from its role as a biomarker in cancer diagnostics and its involvement in embryogenesis. PAX8 interactions with other proteins such as chromatin remodeling complexes make it a valuable target for studying transcriptional regulation mechanisms. Recent studies have demonstrated that PAX8 promotes angiogenesis in ovarian cancer, highlighting its potential as a therapeutic target .
HRP-conjugated PAX8 antibodies are particularly valuable for:
Immunohistochemistry on paraffin-embedded tissues (IHC-P) for direct visualization without secondary antibody steps
Western blot analysis with enhanced sensitivity and reduced background
Flow cytometry applications requiring minimal protocol steps
CyTOF (mass cytometry) experiments
The HRP conjugation eliminates the need for secondary antibody incubation, reducing protocol time and potential non-specific binding. This is especially advantageous when working with limited tissue samples or when performing multiplex staining procedures.
PAX8 exhibits predominantly nuclear localization, consistent with its function as a transcription factor . This localization pattern can be effectively visualized using immunofluorescence or immunohistochemistry techniques.
For optimal visualization:
Use appropriate antigen retrieval methods (heat-mediated antigen retrieval with Tris/EDTA buffer pH 9 has shown good results)
Apply the HRP-conjugated PAX8 antibody at the recommended dilution (typically 1/500-1/1000 for IHC-P)
For immunofluorescence, counterstain nuclei with DAPI to confirm nuclear localization
For IHC-P with HRP-conjugated antibodies, develop with DAB substrate and counterstain with hematoxylin
High-resolution immunofluorescence analysis can reveal the precise nuclear co-localization of PAX8 with other transcription factors such as SOX17, as demonstrated in fallopian tube secretory cell lines and high-grade serous ovarian carcinoma (HGSOC) cell lines .
PAX8 forms large molecular complexes of approximately 600 kDa with various proteins involved in chromatin remodeling and transcriptional regulation . To study these interactions using HRP-conjugated PAX8 antibodies:
Co-immunoprecipitation followed by Western blot analysis:
Prepare nuclear extracts from cells of interest
Perform immunoprecipitation using anti-PAX8 antibodies
Elute the complexes and analyze by Western blot for potential interacting partners
The HRP conjugation can provide direct detection of PAX8 in control blots
Proximity Ligation Assay (PLA):
Size-exclusion chromatography:
Known PAX8 Interacting Proteins | Function | Differential Expression in Cancer vs. Normal Cells |
---|---|---|
CHD4 | Component of NuRD complex | Enriched in cancer cells |
GATAD2A (p66α) | Transcriptional repressor | Enriched in cancer cells |
MTA2 | Metastasis-associated protein | Enriched in cancer cells |
HDAC1 | Histone deacetylase | >100-fold different between normal and cancer cells |
RBBP4 | Retinoblastoma binding protein | >100-fold different between normal and cancer cells |
SOX17 | Transcription factor | Markedly increased in HGSOC compared to FTE cells |
Chromatin immunoprecipitation (ChIP) experiments with PAX8 antibodies require rigorous controls and validation:
Input Controls:
Reserve a portion of the chromatin sample before immunoprecipitation
This represents the starting material and accounts for differences in DNA amounts
Negative Controls:
Positive Controls:
Validation Steps:
Confirm antibody specificity by Western blot prior to ChIP
Verify protein expression in your cell model using immunofluorescence
Optimize chromatin fragmentation to obtain 200-500 bp fragments
Validate ChIP results with independent methodologies (e.g., reporter assays)
Recent advances include ChIC/CUT&RUN-seq applications, which offer improved signal-to-noise ratio compared to traditional ChIP-seq, particularly suitable for certain PAX8 antibody clones like EPR13510 .
Ovarian cancer exhibits significantly higher PAX8 expression compared to normal ovarian tissue, making PAX8 an important marker for high-grade serous ovarian carcinoma (HGSOC) . To optimize PAX8 detection in these samples:
Tissue Processing:
Use formalin-fixed, paraffin-embedded (FFPE) sections of 4-5 μm thickness
Ensure proper fixation time (24 hours) to preserve antigenicity
Antigen Retrieval:
Antibody Selection and Dilution:
Detection Systems:
For HRP-conjugated antibodies, use DAB substrate with optimized development time
Counterstain with hematoxylin for nuclear contrast
Interpretation Guidelines:
Tissue Type | PAX8 Expression | SOX17 Co-expression | Notes |
---|---|---|---|
Normal ovary | Low/Absent | Low/Absent | Not expressed in normal ovarian surface epithelial cells |
Normal fallopian tube | Positive (secretory cells) | Positive (secretory cells) | Co-expression in FT secretory epithelial cells |
Simple ovarian inclusion cysts | Positive | Variable | Used for differential diagnosis |
HGSOC | Strong positive | Strong positive | Abundant expression of both markers |
Mucinous adenocarcinomas | Rarely positive | Variable | PAX8 rarely expressed |
Endometrioid ovarian carcinoma | Frequently positive | Variable | Used for histotype classification |
Clear cell carcinoma | Frequently positive | Variable | Used for histotype classification |
Non-specific background is a common challenge when using HRP-conjugated antibodies in Western blot applications. To minimize this issue:
Blocking Optimization:
Test different blocking agents (5% non-fat milk, 5% BSA, commercial blocking buffers)
Extend blocking time to 1-2 hours at room temperature or overnight at 4°C
Antibody Dilution:
Washing Protocol:
Increase washing time and number of washes (5-6 washes of 5-10 minutes each)
Use TBS-T (0.1% Tween-20) for washing
Reducing Agents:
Ensure complete reduction of samples with fresh DTT or β-mercaptoethanol
This prevents non-specific binding to partially reduced proteins
Detection Sensitivity:
Use highly sensitive ECL substrates appropriate for your expected signal intensity
Short exposure times can help minimize background while capturing specific signals
Western blot analysis has successfully detected PAX8 in various cell lines including HEK-293T and SK-OV-3 using anti-PAX8 antibodies at 1/10000 and 1/1000 dilutions respectively .
PAX8 exists in multiple isoforms due to alternative splicing, which can complicate interpretation of experimental results. To differentiate between these isoforms:
Antibody Selection:
Western Blot Resolution:
Use lower percentage SDS-PAGE gels (8-10%) for better separation of closely spaced bands
Extend running time to achieve maximum separation
Use precision plus protein standards for accurate molecular weight determination
Mass Spectrometry Analysis:
For definitive isoform identification, consider mass spectrometry following immunoprecipitation
This approach can identify specific peptides unique to each isoform
RT-PCR Analysis:
Design primers specific to different exon junctions to detect alternative splicing events
Validate PAX8 isoform expression at the mRNA level before protein analysis
Western blot analysis typically reveals PAX8 as a protein of approximately 62kDa, but closely related isoforms may appear as multiple bands in the 48-62kDa range .
PAX8 plays a crucial role in ovarian carcinogenesis, particularly in high-grade serous ovarian carcinoma (HGSOC). HRP-conjugated PAX8 antibodies can be utilized to explore this process through:
Characterization of PAX8-dependent transcriptional networks:
Analysis of PAX8 protein complexes:
Use biochemical affinity-purification methods to isolate endogenous PAX8 protein complexes
Nuclear extracts from ovarian carcinoma cell lines (OVCAR4, KURAMOCHI, OVSAHO) and immortalized fallopian tube secretory epithelial cells can be compared
Size-exclusion chromatography and mass spectrometry analysis reveal PAX8 exists in approximately 600 kDa complexes with chromatin remodeling components
Visualization of PAX8-SOX17 interactions:
Functional studies of PAX8 in carcinogenesis:
Deploy CRISPR/Cas9 gene editing to modulate PAX8 expression
Assess effects on cell proliferation, migration, invasion, and angiogenesis
Evaluate tumor growth in xenograft models following PAX8 modulation
Multiplexed detection of PAX8 with other cancer biomarkers provides valuable diagnostic and research insights. Here are effective protocols:
Multiplex Immunofluorescence:
Use directly conjugated primary antibodies with different fluorophores
For HRP-conjugated PAX8 antibodies, employ tyramide signal amplification (TSA) with spectrally distinct fluorophores
Include PAX8 with markers such as SOX17, WT1, and p53 for HGSOC characterization
Use spectral imaging and unmixing to resolve overlapping emission spectra
Sequential Immunohistochemistry:
For HRP-conjugated antibodies on the same section:
Perform first staining with PAX8-HRP and develop with DAB (brown)
Strip or block remaining HRP activity
Apply second HRP-conjugated antibody and develop with alternative chromogen (e.g., AEC, red)
Counterstain and analyze
Mass Cytometry (CyTOF):
Multiplexed Protein Arrays:
Digital Spatial Profiling:
Combine PAX8 detection with geographical tissue mapping
Correlate PAX8 expression with other markers in specific microenvironments
This provides insights into tumor heterogeneity and microenvironment interactions
PAX8 represents a promising therapeutic target for cancers where it plays a crucial role in tumorigenesis. HRP-conjugated PAX8 antibodies can facilitate research in developing targeted therapies through:
Identifying druggable protein-protein interactions:
Evaluating PAX8 inhibition strategies:
Screen for small molecule inhibitors that disrupt PAX8 binding to DNA or protein partners
Use HRP-conjugated PAX8 antibodies in high-throughput screening assays
Validate hits with secondary assays including ChIP, co-IP, and cellular phenotypic assays
Developing PAX8-targeted antibody-drug conjugates:
Explore internalizing antibodies against PAX8 for payload delivery
Test efficacy in PAX8-positive cancer models
Evaluate specificity using PAX8 knockout controls
Precision medicine applications:
Stratify patients based on PAX8 expression patterns
Correlate PAX8 complex formation with treatment response
Develop companion diagnostics for PAX8-targeted therapies
PAX8's role in promoting angiogenesis in ovarian cancer suggests that targeting this transcription factor could have anti-angiogenic effects, potentially enhancing current anti-angiogenic therapies.
Despite significant progress, several methodological advances are needed to fully understand PAX8 regulation:
Single-cell analysis of PAX8 expression and function:
Develop protocols for single-cell ChIP-seq to map PAX8 binding in rare cell populations
Combine with single-cell RNA-seq to correlate binding with gene expression
This would reveal cell-to-cell heterogeneity in PAX8 function within tumors
Improved protein-protein interaction detection methods:
Develop more sensitive techniques to detect transient interactions
Implement BioID or APEX proximity labeling with PAX8 to identify the complete interactome
Quantitative interactome analysis between normal and cancer states
Live-cell imaging of PAX8 dynamics:
Develop approaches to visualize PAX8 binding and complex formation in living cells
Engineer cell lines expressing fluorescently tagged PAX8 at endogenous levels
Study the dynamics of PAX8 recruitment to target genes during differentiation or carcinogenesis
Structural studies of PAX8 complexes:
Determine the crystal structure of PAX8 bound to DNA and protein partners
Use cryo-EM to resolve the structure of larger PAX8-containing complexes
This would facilitate structure-based drug design targeting PAX8
Improved antibody technology:
Develop recombinant antibodies with enhanced specificity for different PAX8 isoforms
Create antibodies that selectively recognize post-translationally modified PAX8
Engineer bi-specific antibodies for improved multiplexed detection