RNF39 antibodies are utilized in diverse experimental contexts:
HIV-1 Replication: RNF39 knockdown reduces HIV-1 replication in HEK293T cells by 35% (p = 0.021), while overexpression increases viral replication by 137.9% .
Antiviral Signaling: RNF39 mediates K48-linked ubiquitination of DDX3X, promoting its proteasomal degradation and suppressing RIG-I-like receptor (RLR)-triggered innate immune responses .
Mouse Models: Rnf39-deficient mice exhibit enhanced IFN-β production and reduced RNA viral replication (e.g., VSV) .
Salivary Duct Carcinoma (SDC): RNF39, alongside ADAMTS1 and DSC1, is a prognostic biomarker for aggressive SDC. Anti-RNF39 antibodies enable immunohistochemical stratification of high-risk patients .
Synaptic Plasticity: RNF39 is implicated in long-term potentiation (LTP) via its RING finger domain, as shown in rat hippocampal studies .
RNF39 (Ring Finger Protein 39) is a member of the RING finger protein family characterized by a zinc finger domain that facilitates protein-protein interactions. Recent research has implicated RNF39 as a potential biomarker in several pathological conditions, most notably in salivary duct carcinoma (SDC), where it forms part of a four-gene set (alongside ADAMTS1, DSC1, and IGLL5) for predicting aggressive disease progression. This gene set has emerging value as a predictive biomarker to stratify patients who may benefit from additional systemic or radiation therapies . RNF39's zinc finger domain suggests potential roles in transcriptional regulation, protein ubiquitination, and signal transduction pathways, making it an important target for fundamental research and clinical applications.
RNF39 antibodies are primarily developed in several host species, each offering specific advantages for different research applications. Based on available commercial antibodies, the most common host species include:
Selection of the appropriate host species depends on your experimental design, particularly when conducting multi-labeling experiments where avoiding cross-reactivity is essential .
RNF39 antibodies have been validated for multiple experimental applications, with varying effectiveness depending on the specific antibody clone and format. The primary validated applications include:
Western Blotting (WB): For detecting RNF39 protein in cell or tissue lysates
Immunohistochemistry (IHC): For localization in tissue sections (both paraffin-embedded and frozen)
Immunofluorescence (IF): For subcellular localization studies
Enzyme-Linked Immunosorbent Assay (ELISA): For quantitative detection
Immunocytochemistry (ICC): For cellular localization studies
The application range varies by specific antibody product. For instance, monoclonal antibody clone 5E10 has been validated for WB, IHC, IF, and IHC(p), making it versatile for multiple experimental designs . When selecting an antibody, ensure it has been specifically validated for your intended application to avoid experimental failures.
The choice between monoclonal and polyclonal RNF39 antibodies should be guided by your specific experimental requirements:
Monoclonal RNF39 Antibodies:
Provide high specificity to a single epitope, reducing background
Offer consistent lot-to-lot reproducibility for longitudinal studies
Available clones such as 5E10 and 4D3 target specific regions of RNF39
Best suited for applications requiring high specificity like identifying specific isoforms
Polyclonal RNF39 Antibodies:
Recognize multiple epitopes, potentially increasing detection sensitivity
Better for detecting proteins with post-translational modifications or in denatured states
Available for various regions (N-terminal, C-terminal, and specific amino acid sequences)
Advantageous for applications like IHC where antigen retrieval might alter epitopes
For critical research requiring absolute reproducibility across experiments, monoclonal antibodies like clone 5E10 offer consistent results. For maximum sensitivity in detecting low-abundance RNF39, polyclonal antibodies targeting multiple epitopes may provide better results .
Before incorporating an RNF39 antibody into your research workflow, perform these validation experiments to ensure reliability:
Positive and Negative Controls:
Use cell lines or tissues with known RNF39 expression levels
Include RNF39 knockout or knockdown samples as negative controls
Epitope-Specific Validation:
Cross-Reactivity Assessment:
Application-Specific Validation:
For WB: Confirm molecular weight (expected: approximately 45 kDa)
For IHC/IF: Compare staining patterns with published literature
For quantitative applications: Establish standard curves and determine linear detection range
Antibody Titration:
Perform dilution series to identify optimal working concentration
Assess signal-to-noise ratio across dilutions
Complete validation ensures experimental reproducibility and prevents misleading results from non-specific antibody binding .
For effective immunohistochemical detection of RNF39 in tissue samples, follow this optimized protocol based on published methodologies:
Tissue Preparation:
Fix tissues in 10% neutral buffered formalin for 24-48 hours
Process and embed in paraffin following standard histological procedures
Section tissues at 4-5 μm thickness onto positively charged slides
Antigen Retrieval:
Heat-induced epitope retrieval (HIER) in citrate buffer (pH 6.0) for 20 minutes
Alternative: EDTA buffer (pH 9.0) may provide better results for certain antibody clones
Immunostaining Protocol:
Block endogenous peroxidase with 3% H₂O₂ in methanol (10 minutes)
Block non-specific binding with 5% normal serum from same species as secondary antibody (30 minutes)
Incubate with primary RNF39 antibody (recommended dilutions: 1:100 for monoclonal 5E10, 1:50-1:200 for polyclonal antibodies)
Incubate overnight at 4°C or 1 hour at room temperature
Apply appropriate HRP-conjugated secondary antibody
Develop with DAB substrate and counterstain with hematoxylin
Controls and Validation:
Include positive control tissues (based on the Human Protein Atlas data)
Include negative controls (primary antibody omission and isotype controls)
This protocol has been successfully employed in salivary duct carcinoma research, where RNF39 protein expression was evaluated as a potential prognostic marker .
Successful Western blot detection of RNF39 requires careful optimization of several parameters:
Sample Preparation:
Extract proteins using RIPA buffer supplemented with protease inhibitors
Include phosphatase inhibitors if phosphorylated forms are of interest
Determine optimal protein loading (typically 20-50 μg of total protein)
Gel Electrophoresis and Transfer:
Use 10-12% polyacrylamide gels for optimal resolution of RNF39 (anticipated MW ~45 kDa)
Transfer to PVDF membranes at 100V for 1 hour or 30V overnight at 4°C
Verify transfer efficiency with reversible protein stains before blocking
Antibody Incubation:
Block with 5% non-fat dry milk or BSA in TBST for 1 hour at room temperature
Dilute primary RNF39 antibody according to manufacturer's recommendations (typically 1:500-1:2000)
Incubate with primary antibody overnight at 4°C with gentle agitation
Wash thoroughly (4 × 5 minutes with TBST)
Incubate with appropriate HRP-conjugated secondary antibody (1:5000-1:10000) for 1 hour
Detection and Troubleshooting:
Use enhanced chemiluminescence (ECL) detection system
Start with shorter exposure times (30 seconds) and increase as needed
If background is high, increase washing steps or adjust blocking conditions
If signal is weak, try longer primary antibody incubation or signal amplification systems
This optimized protocol incorporates best practices from published literature on RING finger protein detection and vendor recommendations for specific RNF39 antibodies .
Non-specific binding is a common challenge with antibody-based detection of RNF39. Several factors may contribute to this issue:
Causes of Non-Specific Binding:
Structural Homology: RNF39 belongs to the RING finger protein family, which shares conserved domains with other family members. This structural similarity can lead to cross-reactivity, particularly with polyclonal antibodies.
Antibody Quality: Some commercial antibodies may not undergo rigorous validation against a panel of similar proteins. Antibodies validated only against the immunogen peptide may show cross-reactivity in complex biological samples.
Sample Complexity: Tissue samples, particularly those that are fixed and processed, can present epitopes that bind antibodies non-specifically due to protein modifications or denaturation.
Remediation Strategies:
Epitope-Specific Antibodies: Choose antibodies targeting unique regions of RNF39. The N-terminal region has distinct sequences that can provide greater specificity .
Blocking Optimization: Increase blocking agent concentration (5-10% normal serum or BSA) and duration (up to 2 hours at room temperature).
Antibody Titration: Perform careful dilution series to identify the optimal concentration that maximizes specific signal while minimizing background.
Pre-Absorption Controls: Pre-incubate your antibody with the immunizing peptide when available to confirm specificity of staining patterns.
Alternative Validation: Confirm results using antibodies from different host species or targeting different epitopes of RNF39.
Implementing these strategies will significantly improve the signal-to-noise ratio in your RNF39 detection experiments .
Inconsistent detection of RNF39 across tissue samples can result from both biological variation and technical factors:
Biological Factors:
Expression Level Variability: RNF39 expression may naturally vary across different tissues, cell types, or disease states.
Protein Localization: RNF39 may shuttle between subcellular compartments, affecting epitope accessibility.
Post-Translational Modifications: Modifications may alter epitope recognition in tissue-specific ways.
Technical Solutions:
Antigen Retrieval Optimization:
Test multiple antigen retrieval methods (heat vs. enzymatic)
Compare different buffer systems (citrate pH 6.0 vs. EDTA pH 9.0)
Adjust retrieval duration (10-30 minutes)
Fixation Considerations:
Standardize fixation protocols across specimens
For prospective studies, consider using freshly frozen tissues in parallel
Document fixation duration for retrospective analyses
Signal Amplification:
Implement tyramide signal amplification for low-abundance detection
Consider polymer-based detection systems for increased sensitivity
Multi-Antibody Approach:
Use antibodies targeting different RNF39 epitopes in parallel
Compare results from monoclonal (e.g., clone 5E10) and polyclonal antibodies
Quantitative Controls:
Include standardized positive control tissues in each experiment
Normalize signal intensity against housekeeping proteins
This comprehensive approach has proven effective in standardizing RNF39 detection in heterogeneous samples, such as those used in salivary duct carcinoma studies .
RNF39 antibody has emerged as a valuable tool in cancer research, particularly in the study of salivary duct carcinoma (SDC) and potentially other malignancies:
Salivary Duct Carcinoma Research:
A groundbreaking study by Kohsaka et al. (2022) identified RNF39 as part of a four-gene prognostic signature (alongside ADAMTS1, DSC1, and IGLL5) for aggressive SDC. This research utilized RNF39 antibodies for immunohistochemical validation of expression patterns observed in genomic analyses .
The study demonstrated that:
RNF39 protein expression correlates with specific transcriptional signatures
Immunohistochemical detection of RNF39 can help stratify patients with different prognostic outcomes
RNF39 protein levels may help predict response to adjuvant therapies
Methodological Approaches:
Tissue Microarray Analysis: RNF39 antibodies are used to screen multiple patient samples simultaneously
Correlation with Clinical Outcomes: Expression levels are analyzed in relation to patient survival and treatment response
Multi-Marker Panels: RNF39 staining is often combined with other markers (CD3, ADAMTS1, DSC1) for improved prognostic value
Future Research Directions:
Investigating RNF39's functional role in tumor progression through antibody-based protein interaction studies
Developing RNF39-targeted therapeutics based on structural insights
Expanding RNF39 biomarker validation to other cancer types with similar molecular features
These applications highlight the crucial role of high-quality, validated RNF39 antibodies in translational cancer research .
Investigating RNF39 protein interactions presents several unique challenges that require specialized antibody-based approaches:
Key Challenges:
RING Domain Complexities:
The zinc finger RING domain of RNF39 mediates protein-protein interactions
These interactions may be transient or condition-dependent
Standard immunoprecipitation may disrupt weak or pH-sensitive interactions
Post-Translational Modifications:
Ubiquitination activities associated with RING finger proteins may complicate detection
Modifications may mask epitopes recognized by available antibodies
Conformational States:
RNF39 may adopt different conformations depending on binding partners
Some epitopes may be accessible only in specific conformational states
Advanced Methodological Solutions:
Proximity Ligation Assays (PLA):
Use RNF39 antibodies in combination with antibodies against suspected interacting partners
PLA provides single-molecule resolution of protein interactions in situ
Protocol modifications: Use mild fixation (2% PFA, 10 minutes) and optimize antibody concentrations
Cross-Linking Immunoprecipitation:
Implement protein cross-linking before cell lysis (1-2% formaldehyde, 10 minutes)
Use RNF39 antibodies conjugated to magnetic beads for gentle pull-down
Verify specificity with reciprocal immunoprecipitation using antibodies against interaction partners
Antibody-Based Protein Complementation Assays:
Engineer split reporter proteins fused to anti-RNF39 antibody fragments
Reconstitution occurs when RNF39 interacts with tagged partner proteins
Provides dynamic, real-time monitoring of interactions
These advanced approaches can help overcome the inherent challenges in studying the dynamic protein interaction network of RNF39, providing insights into its functional roles in normal physiology and disease states .
Integrating RNF39 antibody-based data into multi-omics research frameworks provides comprehensive insights into its biological functions and clinical relevance:
Integration Strategies:
Proteogenomic Correlation:
Map RNF39 antibody-detected protein levels against RNA-seq expression data
Identify post-transcriptional regulation mechanisms
Correlation matrix example:
Sample Type | RNF39 Protein (IHC H-score) | RNF39 mRNA (FPKM) | Correlation Coefficient |
---|---|---|---|
Normal Tissue | 10-50 | 5-20 | 0.72 |
Tumor Tissue | 80-200 | 30-120 | 0.63 |
Cell Lines | 40-180 | 15-90 | 0.81 |
Functional Proteomics:
Use RNF39 antibodies for immunoprecipitation followed by mass spectrometry
Map the RNF39 interactome under different cellular conditions
Correlate with phosphoproteomics data to identify signaling networks
Spatial Multi-Omics:
Combine RNF39 immunofluorescence with in situ transcriptomics
Map spatial distribution of RNF39 protein in relation to its transcriptional dependencies
Correlate with metabolomic profiles in tissue microenvironments
Implementation Protocol:
Obtain tissue or cell samples and divide for parallel processing
Process for antibody-based assays (IHC, IF, Western blot) and omics analyses (RNA-seq, proteomics)
Perform RNF39 antibody validation on a subset of samples
Establish normalization procedures across platforms
Apply statistical integration methods (MOFA, DIABLO, mixOmics)
Case Study Application:
In salivary duct carcinoma research, RNF39 antibody data has been successfully integrated with transcriptomic profiles to validate a four-gene prognostic signature. This integration strengthened the clinical relevance of the findings and provided mechanistic insights into the role of RNF39 in disease progression .
Cross-species application of RNF39 antibodies requires careful evaluation of epitope conservation and validation in each target species:
Epitope Conservation Analysis:
RNF39 sequence homology varies across species, with implications for antibody cross-reactivity. For example:
Human-Mouse homology: Varies by region, highest in functional domains
Human-Monkey homology: Generally high across the protein
Validated Cross-Species Applications:
Validation Protocol for Cross-Species Use:
Perform sequence alignment of the antibody's target epitope across species of interest
Test antibody on positive control tissues/cells from each species
Confirm specificity using negative controls (knockout/knockdown where available)
Optimize antibody concentration separately for each species
Document species-specific banding patterns or staining characteristics
Practical Considerations:
For evolutionary studies, consider using antibodies against the most conserved regions of RNF39
For species-specific applications, select antibodies validated in your target organism
When differences are observed across species, verify with multiple antibodies targeting different epitopes
This approach ensures reliable comparative studies while accounting for species-specific variations in RNF39 structure and expression .
Adapting RNF39 antibody protocols for different model organisms requires specific methodological adjustments:
Rodent Models (Mouse/Rat):
Fixation: Reduce fixation time to 12-24 hours for smaller tissues
Antigen retrieval: Increase retrieval time by 5-10 minutes
Primary antibody: Use at 1.5-2× the concentration recommended for human tissues
Background reduction: Include mouse-on-mouse blocking for mouse monoclonal antibodies on mouse tissues
Validation: Compare with RNF39 knockout models when available
Non-Human Primates:
Protocol modifications: Generally minimal due to high sequence homology with human RNF39
Antibody selection: Monoclonal antibody 5E10 shows good cross-reactivity with monkey tissues
Validation: Compare staining patterns with human tissues as reference
Large Animal Models (e.g., Horse):
Tissue processing: Extend dehydration times for larger tissue blocks
Antibody selection: N-terminal antibodies show 92% predicted reactivity with horse RNF39
Optimization: Titrate antibody concentrations specifically for equine tissues
Background reduction: Pre-absorb secondary antibodies with host species proteins
Model-Specific Western Blot Adjustments:
Lysis buffer optimization: Species-specific protease inhibitor cocktails
Loading control selection: Verify housekeeping protein conservation across species
Expected banding patterns: Document species-specific molecular weight variations
These methodological modifications ensure optimal detection of RNF39 across different model organisms while maintaining experimental rigor and reproducibility .