RNF213 is a 591 kDa protein containing a C3HC4-type RING finger domain (E3 ubiquitin ligase activity) and six AAA+ ATPase domains . Antibodies against RNF213 enable detection, localization, and functional studies of this protein in human and animal tissues.
RNF213 antibodies have been critical in elucidating the protein’s roles in immune regulation and pathogen defense:
RNF213 degrades viral proteins via ubiquitin-proteasome pathways. For example:
RNF213 restricts intracellular pathogens like Listeria monocytogenes by promoting nitric oxide (NO) production via DDAH1 regulation .
In Salmonella infections, RNF213 ubiquitinates bacterial LPS through its RZ finger domain .
RNF213 promotes Treg cell differentiation by facilitating K63-linked ubiquitination of PKM2, reducing autoimmune inflammation .
Knockout mice exhibit reduced dendritic cell function and antigen presentation .
Tissue staining:
Cell lines: Detected in HUVEC cells at 1:500–1:1000 dilution .
Knockout validation: Reduced DDAH1 and CYR61 levels in RNF213-depleted macrophages correlate with impaired NO production .
RNF213 (Ring Finger Protein 213) is a large E3 ubiquitin ligase (591 kDa) located in the cytosol and perinuclear space that contains both a C3HC4-type RING finger domain and six AAA+ ATPase domains, making it unique as the only known protein with both these functional domains. RNF213 is critical to study because it plays multifunctional roles in angiogenesis, lipid metabolism, and cell-autonomous immunity. The protein mediates ubiquitination of both proteins (e.g., FLNA and NFATC2) and lipids (e.g., bacterial lipopolysaccharide), indicating its involvement in diverse cellular processes. Additionally, RNF213 has been strongly associated with Moyamoya disease, a rare cerebrovascular disorder, making it an important target for investigating vascular pathologies .
For proper study of RNF213, antibody-based techniques are essential because they allow specific detection of this large protein in various experimental contexts including immunoblotting, immunohistochemistry, and immunofluorescence. These techniques enable researchers to observe native expression patterns, subcellular localization, protein-protein interactions, and changes in expression levels during disease states or in response to various stimuli.
When selecting an RNF213 antibody for research, consider:
Epitope location: Choose antibodies that target conserved regions of the protein, particularly if studying specific domains such as the RING finger or AAA+ ATPase domains.
Validation status: Select antibodies validated for your specific application (Western blot, IHC, IF, IP). RNF213 antibodies have been successfully used in various applications including immunoblotting, immunohistochemistry, and immunofluorescence .
Species reactivity: Ensure the antibody recognizes RNF213 in your experimental species. Human RNF213 has the UniProt ID Q63HN8 .
Clonality: Polyclonal antibodies may offer higher sensitivity by recognizing multiple epitopes, while monoclonal antibodies provide higher specificity and consistency between batches.
Size detection capability: Since RNF213 is a large protein (591 kDa), confirm the antibody can detect proteins in this high molecular weight range, which may require specialized gel systems and transfer protocols.
Recognition of variants: Verify whether the antibody can detect known splice variants or mutant forms, especially when studying Moyamoya disease-associated variants.
Background signal: Review literature or validation data to assess non-specific binding potential, which is particularly important when working with complex tissue samples.
Published validation: Prioritize antibodies cited in peer-reviewed publications that demonstrate successful applications similar to your planned experiments.
When using RNF213 antibodies, the following controls are essential:
Positive control: Include lysates from cells known to express RNF213 (such as endothelial cells or immune cells) to confirm antibody functionality.
Negative control: Use appropriate RNF213-knockout or knockdown samples. Since complete knockout models may be challenging due to RNF213's role in development, siRNA or shRNA knockdown samples serve as good alternatives for antibody validation.
Secondary-only control: Perform parallel experiments omitting the primary RNF213 antibody to identify any non-specific binding from the secondary antibody.
Blocking peptide control: When available, pre-incubate the antibody with its immunizing peptide to demonstrate binding specificity.
Loading control: Include detection of housekeeping proteins (β-actin, GAPDH, tubulin) to normalize RNF213 expression levels across samples.
Titration series: Establish optimal antibody concentration by testing a range of dilutions to determine the concentration that provides the best signal-to-noise ratio.
Cross-reactivity assessment: In multi-protein detection experiments, include controls to ensure the RNF213 antibody doesn't cross-react with other E3 ligases, especially those with similar RING finger domains.
Isotype control: For immunoprecipitation or flow cytometry applications, include an isotype-matched irrelevant antibody control.
Optimized Western Blotting Protocol for RNF213 Detection:
Sample preparation:
Lyse cells in RIPA buffer supplemented with protease inhibitors and phosphatase inhibitors
Include 1-2 mM N-ethylmaleimide to preserve ubiquitination states
Sonicate briefly to shear DNA and reduce sample viscosity
Centrifuge at 14,000 × g for 15 minutes at 4°C to clear lysates
Protein separation:
Use low percentage (4-6%) SDS-PAGE gels or gradient gels (4-12%) to effectively resolve the 591 kDa RNF213 protein
Load 50-100 μg of total protein per lane
Run at lower voltage (80-100V) for longer duration to improve separation of high molecular weight proteins
Transfer:
Employ wet transfer systems with chilled transfer buffer containing 0.05-0.1% SDS to aid transfer of large proteins
Use PVDF membrane with 0.45 μm pore size
Transfer at 30V overnight at 4°C or use a high-molecular-weight transfer protocol
Blocking and antibody incubation:
Block with 5% non-fat dry milk or BSA in TBST for 1-2 hours at room temperature
Incubate with primary RNF213 antibody (typically 1:500-1:2000 dilution) overnight at 4°C
Wash extensively with TBST (4-5 washes, 5-10 minutes each)
Incubate with secondary antibody (typically 1:5000-1:10000) for 1 hour at room temperature
Detection:
Use enhanced chemiluminescence (ECL) detection systems with extended exposure times
For more sensitive detection, consider using fluorescent secondary antibodies and imaging systems
Special considerations:
Confirm complete transfer of high molecular weight proteins using reversible staining methods
Consider using a reference marker specifically for high molecular weight proteins
Optimize exposure time to avoid saturation while maintaining sensitivity
Optimized Immunofluorescence Protocol for RNF213 Subcellular Localization:
Cell preparation:
Culture cells on glass coverslips or chamber slides coated with appropriate substrates
Consider using cells with known RNF213 expression (e.g., endothelial cells)
Fixation options:
Primary method: 4% paraformaldehyde for 15 minutes at room temperature
Alternative method: Methanol fixation (100% methanol at -20°C for 10 minutes) may better preserve certain epitopes
Perform comparative analysis to determine optimal fixation for your specific RNF213 antibody
Permeabilization:
Use 0.2% Triton X-100 in PBS for 10 minutes at room temperature
For gentler permeabilization, consider 0.1% saponin, which may better preserve membrane structures
Blocking:
Block with 3-5% normal serum (matching the species of secondary antibody) with 0.1% BSA in PBS for 1 hour
Antibody incubation:
Dilute primary RNF213 antibody (typically 1:100-1:500) in blocking buffer
Incubate overnight at 4°C in a humidified chamber
Wash 3-5 times with PBS
Incubate with fluorophore-conjugated secondary antibody (1:500-1:1000) for 1 hour at room temperature in the dark
Co-staining considerations:
For co-localization studies, combine with markers for:
Perinuclear region: Nuclear envelope markers (Lamin A/C)
Cytosolic compartments: Specific organelle markers based on research question
For angiogenesis studies: Endothelial markers (CD31, VE-cadherin)
For immune function studies: Combine with bacterial or LPS markers
Nuclear counterstaining:
Counterstain with DAPI or Hoechst (1:5000) for 5 minutes
Mount with anti-fade mounting medium
Imaging recommendations:
Use confocal microscopy for precise subcellular localization
Capture Z-stacks to analyze the full cell volume
Include multi-channel imaging to assess co-localization with other cellular components
Controls:
Include secondary-only control
Include RNF213 knockdown cells as negative control
RNF213 has been reported to localize primarily in the cytosol and perinuclear space , so particular attention should be paid to these regions when analyzing subcellular distribution patterns.
Protocol for Measuring RNF213 E3 Ligase Activity:
Immunoprecipitation-based ubiquitination assay:
a) Cell lysate preparation:
Lyse cells in denaturing buffer (1% SDS, 50 mM Tris-HCl pH 7.5, 0.5 mM EDTA, 1 mM DTT)
Heat at 95°C for 10 minutes to disrupt protein interactions
Dilute 10-fold with non-denaturing buffer (0.5% NP-40, 50 mM Tris-HCl pH 7.5, 150 mM NaCl, 5 mM EDTA)
Include protease inhibitors, phosphatase inhibitors, and deubiquitinase inhibitors (10 mM N-ethylmaleimide)
b) Immunoprecipitation:
Pre-clear lysate with Protein A/G beads
Immunoprecipitate RNF213 using validated antibodies coupled to Protein A/G beads
Wash extensively with high-salt buffer followed by standard wash buffer
c) Ubiquitination detection:
Analyze by SDS-PAGE and immunoblot
Probe with anti-ubiquitin antibody to detect ubiquitinated proteins
Re-probe with anti-RNF213 to confirm successful immunoprecipitation
In vitro ubiquitination assay:
a) Protein purification:
Purify recombinant RNF213 or immunoprecipitate from cell lysates
Verify purity by SDS-PAGE
b) Ubiquitination reaction:
Combine purified RNF213 with E1 enzyme, E2 enzyme, ubiquitin, ATP, and substrate
Incubate at 37°C for 1-2 hours
c) Activity detection:
Analyze reaction products by SDS-PAGE and immunoblotting
Detect ubiquitinated products using anti-ubiquitin antibody
Alternatively, use fluorescently labeled ubiquitin for direct visualization
Cellular ubiquitination assay:
a) Cell preparation:
Transfect cells with tagged ubiquitin (HA-Ub or His-Ub)
Optionally co-transfect with RNF213 wild-type or mutant constructs
For bacterial studies, infect cells with appropriate pathogens (e.g., Salmonella)
b) Enrichment of ubiquitinated proteins:
For His-Ub: perform Ni-NTA pulldown under denaturing conditions
For HA-Ub: perform anti-HA immunoprecipitation
c) Detection:
Controls and validation:
Include catalytically inactive RNF213 mutant (RING domain mutant)
Include reactions without ATP as negative control
For pathogen studies, include both wild-type bacteria and relevant bacterial mutants
Methodological Approaches for Studying Moyamoya Disease Using RNF213 Antibodies:
Patient-derived sample analysis:
Immunohistochemistry (IHC) on vascular tissue from MMD patients compared to controls
Western blot analysis of RNF213 expression levels in peripheral blood mononuclear cells (PBMCs) from patients with different RNF213 genotypes
Correlation of RNF213 protein levels with disease severity and progression
Functional studies with disease-associated variants:
Generate cell lines expressing wild-type RNF213 or MMD-associated variants (especially p.R4810K)
Compare protein stability, subcellular localization, and E3 ligase activity between wild-type and mutant RNF213
Assess differences in substrate recognition and ubiquitination patterns
Vascular remodeling investigation:
Use RNF213 antibodies in combination with endothelial markers in patient-derived induced pluripotent stem cells (iPSCs) differentiated into vascular cells
Track RNF213 expression during angiogenesis assays comparing wild-type and mutant conditions
Analyze RNF213's role in the non-canonical Wnt signaling pathway by monitoring ubiquitination and degradation of FLNA and NFATC2 during vessel regression
Co-immunoprecipitation studies:
Identify RNF213-interacting proteins in vascular tissues from normal vs. MMD patients
Compare protein interaction networks between wild-type and mutant RNF213
Investigate differences in complex formation and substrate binding
Imaging applications:
Use high-resolution confocal microscopy with RNF213 antibodies to analyze vascular smooth muscle cells and endothelial cells from patient samples
Implement proximity ligation assays to detect RNF213 interaction with potential binding partners in situ
Apply super-resolution microscopy techniques to analyze fine details of RNF213 localization in the perinuclear region
RNF213 Variant | Association with MMD | Protein Expression | Suggested Antibody Method |
---|---|---|---|
Wild-type | Reference | Normal | Western blot, IHC, IF |
p.R4810K | Strong association in East Asian populations | Potentially altered stability/localization | Compare protein levels by Western blot; analyze localization by IF |
p.D4013N | Reported in European/American patients | Potentially altered function | Assess E3 ligase activity by ubiquitination assays |
p.A5021V | Familial MMD cases | Potentially altered ATPase activity | Analyze protein interactions by co-IP |
Inflammatory response assessment:
Given RNF213's role in immunity, analyze inflammatory signaling pathways in patient-derived cells with MMD-associated mutations
Evaluate RNF213 expression in response to inflammatory stimuli in cells with wild-type vs. mutant RNF213
Methodological Approaches for Studying RNF213's Antimicrobial Function:
Infection models:
Monitor RNF213 expression and localization during bacterial infection using immunofluorescence
Track recruitment of RNF213 to bacterial entry sites or cytosolic bacteria (particularly for Salmonella or Shigella)
Analyze co-localization with bacterial markers and autophagic machinery
LPS ubiquitination detection:
Use co-immunoprecipitation with RNF213 antibodies followed by detection with anti-LPS antibodies
Implement proximity ligation assays to visualize RNF213-LPS interactions in situ
Develop a three-color immunofluorescence approach to simultaneously visualize RNF213, ubiquitin, and LPS
Bacterial subversion mechanism studies:
Antibacterial autophagy pathway:
Assess the role of RNF213 in antibacterial autophagy by monitoring LC3 recruitment to bacteria
Compare autophagy induction between wild-type and RNF213-depleted cells using confocal microscopy
Investigate the relationship between RNF213-mediated LPS ubiquitination and downstream autophagy activation
LUBAC complex interaction:
Infection Model | Key RNF213 Function | Antibody-Based Detection Method |
---|---|---|
Salmonella infection | LPS ubiquitination; antibacterial autophagy | IF for RNF213 localization to bacteria; Co-IP for RNF213-LPS interaction |
Shigella infection | Target of bacterial effector IpaH1.4 | Western blot to monitor RNF213 degradation; IF to visualize IpaH1.4-RNF213 interaction |
Viral infection | Antiviral immunity mechanisms | Western blot to assess expression changes; IF for subcellular redistribution |
Parasite infection | Immune response to parasites | IF to track localization during parasite invasion |
RNF213 expression regulation:
Analyze RNF213 protein levels in response to different microbial stimuli and interferon treatment
Compare expression patterns in different immune cell types using flow cytometry or immunohistochemistry
Investigate post-translational modifications of RNF213 during infection
Methodological Approaches for Studying RNF213's Function in Lipid Metabolism:
Lipid droplet association studies:
Use co-immunofluorescence to analyze RNF213 localization relative to lipid droplets (stained with BODIPY or similar markers)
Implement super-resolution microscopy to determine precise spatial relationships
Examine changes in localization under different metabolic conditions (normal vs. high fat)
Regulation of lipolysis:
Fatty acid desaturation pathway:
Lipotoxicity protection mechanisms:
Assess how RNF213 regulates lipotoxicity through inhibition of fatty acid desaturation
Analyze cell survival and ER stress markers in relation to RNF213 expression levels
Use immunoprecipitation to identify ubiquitinated targets in the lipid metabolic pathway
Metabolic disease models:
Apply RNF213 antibodies in tissues from metabolic disease models (obesity, diabetes)
Compare RNF213 expression patterns between healthy and diseased states
Correlate RNF213 levels with lipid accumulation and metabolic parameters
Metabolic Condition | RNF213 Function | Suggested Antibody Application |
---|---|---|
Normal metabolism | Baseline regulation | Western blot for expression level; IF for normal localization |
Lipid overload | Inhibition of lipolysis; Lipid droplet formation | IF to track association with lipid droplets; Co-IP to identify binding partners |
Fatty acid stress | Inhibition of desaturation | Western blot to monitor expression changes; IP-MS to identify regulated targets |
Metabolic disease | Potential dysregulation | IHC in affected tissues; Expression correlation with disease markers |
Methodological Approach for Domain-Specific RNF213 Analysis:
The large size of RNF213 (591 kDa) with its multiple functional domains presents unique opportunities for domain-specific investigations. RNF213 contains a RING-type zinc finger domain responsible for E3 ligase activity and six AAA+ ATPase domains . Domain-specific antibodies can provide insights into:
Domain-specific antibody selection and validation:
Choose antibodies targeting specific epitopes within:
RING domain (for investigating E3 ligase function)
AAA+ ATPase domains (for investigating ATP-dependent functions)
RZ finger domain (important for LPS ubiquitination)
Validate specificity using:
Domain deletion constructs
Point mutants in specific domains
Peptide competition assays with domain-specific peptides
Functional domain analysis applications:
Use domain-specific antibodies to:
Track conformational changes upon ATP binding/hydrolysis
Identify which domains interact with specific binding partners
Determine accessibility of domains in different cellular contexts
Domain-specific Co-IP strategy:
Implement a strategy using domain-specific antibodies to:
Pull down RNF213 via different domains
Compare interactome differences when targeting different domains
Identify domain-specific post-translational modifications
Structural biology applications:
Use domain-specific antibodies as tools for:
Cryo-EM studies to stabilize specific conformations
Domain accessibility mapping under different conditions
Epitope mapping to reveal structural features
RNF213 Domain | Function | Antibody Application | Expected Insights |
---|---|---|---|
RING domain | E3 ubiquitin ligase activity | IP followed by ubiquitination assays; IF for substrate co-localization | Regulation of catalytic activity; substrate recognition |
AAA+ ATPase domains | ATP hydrolysis; conformational changes | Conformation-specific antibodies; structural studies | Energy-dependent structural changes; oligomerization state |
RZ finger domain | LPS ubiquitination | Detection during bacterial infection; mutational analysis | Mechanisms of pathogen recognition; binding specificity |
Disease-associated variant analysis:
Apply domain-specific antibodies to:
Determine how MMD-associated mutations affect specific domain functions
Compare domain accessibility between wild-type and mutant proteins
Analyze domain-specific interactions in patient-derived samples
Advanced Methods for Detecting RNF213-Substrate Interactions:
Capturing the often transient interactions between E3 ligases and their substrates presents significant technical challenges, particularly for a large protein like RNF213. The following methodological approaches can help researchers overcome these challenges:
Substrate trapping approaches:
Generate catalytically inactive RNF213 mutants (RING domain mutations) that can bind but not ubiquitinate substrates, prolonging the interaction
Use protein crosslinking methods (e.g., formaldehyde, DSP, or photo-crosslinkers) to stabilize transient interactions
Implement BioID or TurboID proximity labeling fused to RNF213 to identify proteins in close proximity
Real-time interaction monitoring:
Develop FRET-based assays with fluorescently labeled RNF213 and potential substrates
Use bioluminescence resonance energy transfer (BRET) for live-cell monitoring of interactions
Implement single-molecule tracking microscopy to visualize interaction dynamics
Modified immunoprecipitation strategies:
Perform tandem affinity purification with RNF213 antibodies under carefully optimized conditions
Use rapid immunoprecipitation techniques to capture short-lived complexes
Include ATP in buffers to stabilize AAA+ domain-dependent interactions
In situ visualization techniques:
Apply proximity ligation assays (PLA) to visualize RNF213-substrate interactions in fixed cells
Implement split-GFP complementation assays for live-cell visualization
Use super-resolution microscopy techniques to detect co-localization at nanometer resolution
Structural biology approaches:
Generate antibody fragments (Fabs) that can stabilize RNF213-substrate complexes for structural studies
Use cryo-electron microscopy with domain-specific antibodies to visualize substrate binding modes
Implement hydrogen-deuterium exchange mass spectrometry to map interaction interfaces
Substrate Type | Interaction Challenge | Recommended Method | Expected Outcome |
---|---|---|---|
Protein substrates (FLNA, NFATC2) | Rapid ubiquitination and release | Catalytically inactive mutants; crosslinking; PLA | Identification of binding interfaces; temporal dynamics |
LPS (bacterial lipopolysaccharide) | Complex biochemistry; microbial context | BioID proximity labeling; subcellular fractionation | Understanding of interaction in infection context |
Novel substrates | Unknown identity | IP-MS with quantitative proteomics; ubiquitinome analysis | Discovery of new RNF213 targets |
Advanced Validation Strategies for RNF213 Antibodies:
Validating antibody specificity for a large, complex protein like RNF213 (591 kDa) presents significant challenges, particularly in diverse experimental conditions and tissue types. The following comprehensive approach ensures reliable detection:
Genetic validation approaches:
Use CRISPR/Cas9-engineered RNF213 knockout cells as definitive negative controls
Implement siRNA/shRNA knockdown with titration to create a gradient of expression for sensitivity assessment
Generate epitope-tagged RNF213 knock-in cell lines for parallel detection with anti-tag and anti-RNF213 antibodies
For tissues, use conditional knockout models if complete knockout is not viable
Biochemical validation methods:
Perform peptide competition assays with the immunizing peptide
Validate using recombinant protein fragments spanning different RNF213 domains
Implement multiple antibodies targeting different epitopes to confirm specificity
Use mass spectrometry to confirm the identity of immunoprecipitated proteins
Tissue-specific validation:
Include tissue-specific controls (e.g., brain vasculature for Moyamoya studies)
Compare staining patterns across multiple tissue types with known RNF213 expression patterns
Employ RNA-protein correlation using parallel RNA-seq and protein detection
Use RNAscope or similar in situ hybridization techniques alongside immunohistochemistry
Technical optimization for challenging conditions:
Systematically compare fixation methods (paraformaldehyde, methanol, acetone)
Test multiple antigen retrieval protocols for formalin-fixed tissues
Optimize signal amplification methods for low-abundance detection
Establish titration curves across different experimental conditions
Advanced imaging validation:
Use super-resolution microscopy to confirm expected subcellular localization patterns
Implement spectral imaging to distinguish true signal from autofluorescence
Perform co-localization studies with known interaction partners
Use tissue clearing techniques for 3D validation in thick tissue samples
Common Challenges and Solutions for RNF213 Detection:
High molecular weight detection issues:
Challenge: RNF213's large size (591 kDa) makes it difficult to resolve and transfer in standard Western blotting protocols.
Solutions:
Use low percentage gels (3-6%) or gradient gels (4-12%)
Implement specialized transfer techniques:
Extended transfer times (overnight at 30V)
Add 0.05% SDS to transfer buffer
Use discontinued or pulsed-field electrophoresis
Validate complete transfer with reversible staining methods
Consider using capillary-based Western systems for improved transfer of high MW proteins
Low endogenous expression levels:
Challenge: Detecting native RNF213 in many cell types can be difficult due to relatively low expression.
Solutions:
Use signal enhancement methods:
HRP-conjugated polymers instead of standard secondary antibodies
Tyramide signal amplification for immunohistochemistry
Extended exposure times for Western blots
Concentrate protein by immunoprecipitation before analysis
Induce expression with interferon treatment before analysis
Identify cells with higher endogenous expression as positive controls
Non-specific binding and background:
Challenge: Large proteins often show non-specific binding patterns or cross-reactivity.
Solutions:
Optimize blocking conditions:
Test different blocking agents (milk, BSA, normal serum)
Increase blocking time or concentration
Increase washing stringency:
More frequent washes
Higher salt concentration in wash buffers
Pre-adsorb antibodies with cell lysates from RNF213 knockout cells
Use monoclonal antibodies for higher specificity when available
Fixation and epitope accessibility issues:
Challenge: Different fixation methods can affect epitope accessibility.
Solutions:
Compare multiple fixation methods:
Paraformaldehyde (2-4%)
Methanol/acetone
Glyoxal-based fixatives
Optimize antigen retrieval:
Heat-induced epitope retrieval
Enzymatic retrieval
pH-controlled buffers
Use permeabilization optimization:
Test different detergents (Triton X-100, saponin, digitonin)
Vary permeabilization time and concentration
Post-translational modifications altering detection:
Challenge: Ubiquitination or other modifications may mask epitopes or alter migration patterns.
Solutions:
Include deubiquitinases inhibitors in lysis buffers
Test antibodies targeting different domains/epitopes
Use denaturing conditions in sample preparation
Consider phosphatase treatment to eliminate charge-based mobility shifts
Challenge | Technical Solution | Validation Approach |
---|---|---|
High MW detection | Gradient gels; optimized transfer | Confirm with size markers; verify transfer efficiency |
Low expression | Signal amplification; concentration methods | Quantify signal-to-noise ratio; compare to overexpression control |
Background signal | Optimized blocking; increased washing | Secondary-only controls; comparison across multiple antibodies |
Fixation issues | Multiple fixation comparison | Side-by-side processing of control samples |
Post-translational masking | Denaturing conditions; multiple epitope targeting | Treatment with modification-removing enzymes |
Strategies for Resolving Conflicting RNF213 Antibody Data:
Researchers frequently encounter situations where different antibodies against RNF213 produce seemingly contradictory results. This comprehensive framework helps systematically address such discrepancies:
Epitope mapping and comparison:
Determine the exact epitopes recognized by each antibody
Assess whether epitopes might be differentially affected by:
Protein conformation changes due to experimental conditions
Post-translational modifications masking specific regions
Domain-specific interactions with binding partners
Consider whether antibodies recognize different splice variants or isoforms of RNF213
Validation hierarchy establishment:
Create a validation priority framework:
Assign higher confidence to results confirmed by genetic approaches (KO/KD)
Prioritize antibodies with extensive published validation
Consider monoclonal antibodies more reliable for specific applications
Weigh results from multiple detection methods (e.g., IF confirmed by WB)
Implement orthogonal detection methods to break ties between conflicting antibodies
Technical parameter analysis:
Systematically evaluate whether discrepancies arise from:
Different fixation or extraction methods affecting epitope accessibility
Variations in antibody concentration or incubation conditions
Differential sensitivity to denaturation or native conditions
Batch-to-batch variability in polyclonal antibodies
Create standardized protocols to directly compare antibodies under identical conditions
Application-specific optimization:
Recognize that different antibodies may perform optimally in different applications:
Some antibodies work well for Western blot but poorly for IF
Others may excel in native IP but fail in denaturing conditions
Develop application-specific validation criteria rather than expecting universal performance
Biological context consideration:
Assess whether conflicting results reflect actual biological variability:
Cell type-specific post-translational modifications
Context-dependent protein interactions masking epitopes
Different subcellular pools of RNF213 with distinct properties
Disease-specific alterations in protein structure or localization
Conflicting Observation | Investigation Approach | Resolution Strategy |
---|---|---|
Different subcellular localization patterns | Multi-antibody IF with domain-specific antibodies | Determine if different domains localize differently or if certain epitopes are masked in specific compartments |
Varying molecular weight detection | Gradient gels with multiple antibodies and loading controls | Identify potential splice variants, degradation products, or post-translational modifications |
Discrepant expression levels across tissues | Parallel RNA-seq and protein analysis; multiple antibody validation | Establish tissue-specific expression profiles with orthogonal validation |
Contradictory interaction partners | Domain-specific antibodies for IP; crosslinking approaches | Map domain-specific interactions and context-dependent binding |
Inconsistent disease-associated changes | Patient-derived samples with multiple antibodies; genetic validation | Identify disease-specific modifications or conformational changes affecting epitope accessibility |
Advanced Methods for Specific RNF213 Detection Among RING Proteins:
The human genome encodes hundreds of RING finger proteins with structural similarities to the RING domain of RNF213, creating potential specificity challenges. The following comprehensive approach ensures specific detection:
Unique domain combination exploitation:
Target detection strategies to RNF213's unique combination of domains:
No other protein combines RING domains with six AAA+ ATPase domains
Design antibodies targeting the junction regions between these domains
Use the unique RZ finger domain involved in LPS ubiquitination for specific detection
Implement sandwich assays requiring recognition of multiple unique domains
Size-based differentiation:
Leverage RNF213's exceptional size (591 kDa):
Use size exclusion chromatography before immunodetection
Implement specialized gel systems optimized for very large proteins
Apply gradient gels that clearly separate high molecular weight proteins
Combine molecular weight determination with specific epitope detection
Sequence divergence targeting:
Focus antibody development on regions with lowest homology to other RING proteins:
Target non-conserved loops or linker regions
Avoid highly conserved zinc-coordinating residues in antibody design
Develop peptide-specific antibodies against unique RNF213 sequences
Validate specificity against closely related RING finger proteins
Functional discrimination approaches:
Exploit RNF213's unique functional characteristics:
Analyze ATP-dependent activities not present in most RING proteins
Target RNF213's distinctive ability to ubiquitinate lipids like LPS
Focus on unique substrate specificities (FLNA, NFATC2)
Implement activity-based probes that selectively label active RNF213
Advanced purification and detection:
Use tandem purification strategies:
Sequential immunoprecipitation with antibodies against different domains
Combine size-based separation with immunoaffinity purification
Implement substrate-based affinity capture for functional RNF213
Apply mass spectrometry for definitive identification after enrichment
Similar RING Protein | Distinguishing Feature | Recommended Approach | Validation Method |
---|---|---|---|
Other E3 ubiquitin ligases | Size; domain arrangement; ATPase activity | Size-based separation; domain-specific antibodies | MS confirmation; activity assays |
RING-domain only proteins | Presence of AAA+ domains; molecular weight | Target non-RING regions; use molecular weight | Knockout controls; recombinant protein standards |
RING proteins with ATPase function | Specific domain organization; substrate specificity | Target unique domain junctions; use substrate-based detection | Competitive binding assays; domain deletion controls |
RNF protein family members | Size; specific amino acid sequences | Target non-conserved regions; use size discrimination | Multi-epitope validation; specific knockout controls |