The RNF103 antibody is a specialized immunoglobulin designed to target RNF103 (RING Finger Protein 103), an E3 ubiquitin ligase critical for protein degradation and cellular regulation. RNF103 contains a RING-H2 domain, enabling its role in ubiquitination processes, and is implicated in neuronal function, stress responses, and disease pathology. This antibody serves as a research tool for studying RNF103’s biological roles, including its involvement in neurodegeneration, cancer, and metabolic disorders.
RNF103 antibodies are generated using recombinant protein immunogens and validated for specificity. Below are key variants:
Clone 3E7 (Sigma-Aldrich):
Rabbit Polyclonal (HPA057922, Sigma-Aldrich):
RNF103 antibodies are instrumental in studying its role in:
Alzheimer’s Disease: RNF103 interacts with Derlin-1 and VCP, suggesting involvement in ERAD pathways. Overexpression in Alzheimer’s brains may regulate neuronal homeostasis .
Stress Responses: Induced by ECT and antidepressants, linking RNF103 to synaptic plasticity and mood regulation .
Pancreatic Adenocarcinoma: Overexpression correlates with invasive potential and MMP-9 activity .
Ovarian and Melanoma: Identified as a potential biomarker in autoantibody panels .
Bile Acid Transport: RNF103 ubiquitinates mutant bile salt pumps (BSEP/ABCB11), linking it to cholestatic liver diseases .
Alzheimer’s: RNF103 mRNA is elevated in the frontal cortex of Alzheimer’s patients, correlating with neuronal dysfunction .
Anxiety Models: Kf-1 knockout mice exhibit increased anxiety, implicating RNF103 in emotional regulation .
Antibody Engineering: Bispecific antibodies pairing RNF103 with transmembrane receptors (e.g., IGF1R) enable targeted degradation. This approach leverages RNF103’s E3 ligase activity to modulate receptor signaling .
Specificity: Cross-reactivity with related RING finger proteins (e.g., RNF43, ZNRF3) requires rigorous validation .
Therapeutic Potential: RNF103-based PROTABs (proteolysis-targeting antibodies) may offer novel strategies for degrading oncogenic receptors .
Biomarker Utility: RNF103 autoantibodies in cancer and neurodegeneration warrant further validation in clinical cohorts .
RNF103 (Ring Finger Protein 103, also known as KF-1, HKF-1, or ZFP-103) is an E3 ubiquitin-protein ligase that functions in the endoplasmic reticulum-associated protein degradation (ERAD) pathway. This protein contains a RING-H2 finger motif involved in protein-protein and protein-DNA interactions . RNF103 is particularly significant in neuroscience research as it shows high expression in the cerebellum and its expression in the frontal cortex and hippocampus can be induced by electroconvulsive treatment and chronic antidepressant treatment, suggesting a potential role in depression mechanisms . Studying RNF103 can provide insights into protein quality control, neurological disorders, and cellular stress responses.
Currently, researchers can access several types of RNF103 antibodies, with the most common being rabbit polyclonal antibodies. These antibodies typically target specific regions of the RNF103 protein, such as the 243-293 amino acid region . The antibodies are generally unconjugated and affinity-purified, making them suitable for various applications. Both commercial sources and custom antibody services provide RNF103 antibodies with validated reactivity against human, mouse, and rat samples .
RNF103 antibodies have been validated for several research applications, primarily:
Western Blot (WB): For detecting and quantifying RNF103 protein in cell or tissue lysates. Recommended dilution ranges are typically 1:500-2000 .
Immunohistochemistry (IHC): For visualizing RNF103 distribution in tissue sections. Recommended dilution ranges are typically 1:50-300 for IHC-P (paraffin-embedded tissues) .
Some antibodies may also be suitable for additional applications such as immunofluorescence, immunoprecipitation, or ELISA, though specific validation for these applications should be confirmed with the antibody manufacturer.
For optimal preservation of RNF103 antibody activity, researchers should follow these storage guidelines:
Avoid repeated freeze-thaw cycles, which can lead to antibody degradation and loss of binding efficiency .
The antibodies are typically provided in a stabilizing solution of PBS containing 50% Glycerol, 0.5% BSA, and 0.02% Sodium Azide .
For working aliquots, small volumes can be maintained at 4°C for up to one month, but long-term storage should remain at -20°C.
When designing experiments with RNF103 antibodies, researchers should include the following controls:
Positive control: Tissues or cell lines known to express RNF103 (e.g., cerebellum tissue) .
Negative control: Tissues or cell lines with low or no expression of RNF103, or RNF103 knockout samples if available.
Secondary antibody control: Samples treated with only the secondary antibody to identify any non-specific binding.
Isotype control: Using a non-specific IgG from the same species as the primary antibody to identify potential non-specific interactions.
Blocking peptide control: If available, pre-incubating the antibody with the immunogen peptide to confirm specificity.
Optimizing RNF103 antibody performance for Western blot requires careful attention to several parameters:
| Parameter | Recommendation | Rationale |
|---|---|---|
| Lysate preparation | Include protease inhibitors; use RIPA buffer with 1% SDS | RNF103 is a membrane protein; stronger detergents help solubilization |
| Protein loading | 20-50 μg of total protein | Ensures adequate detection while minimizing background |
| Blocking solution | 5% non-fat milk in TBST or 3% BSA in TBST | Reduces non-specific binding |
| Primary antibody dilution | Start with 1:1000, optimize from 1:500-2000 | Balance between signal strength and background |
| Incubation time/temperature | Overnight at 4°C | Improves specific binding while reducing background |
| Washing steps | 3-5 washes with TBST, 5-10 minutes each | Removes unbound antibody, reducing background |
| Secondary antibody | HRP-conjugated anti-rabbit IgG at 1:5000-10000 | Provides sensitive detection with minimal background |
Additionally, for membrane proteins like RNF103, avoiding boiling your samples can help prevent protein aggregation - instead, incubate at 37°C for 30 minutes in Laemmli buffer. If signal strength is an issue, consider using a more sensitive detection method such as ECL Plus or Super Signal West Femto .
Successful immunohistochemistry with RNF103 antibodies requires optimization of several critical parameters:
Antigen retrieval: As RNF103 is an endoplasmic reticulum membrane protein, heat-induced epitope retrieval using citrate buffer (pH 6.0) or EDTA buffer (pH 9.0) is often necessary. Test both methods to determine optimal conditions.
Antibody dilution: Begin testing at the recommended range (1:50-300 for IHC-P) , then adjust based on signal-to-noise ratio.
Incubation conditions: Longer incubations (overnight at 4°C) typically produce better results than short incubations at room temperature.
Detection system: For low abundance proteins like RNF103, amplification systems such as tyramide signal amplification may improve sensitivity.
Tissue fixation: Overfixation can mask epitopes; standardize fixation protocols (10% neutral buffered formalin for 24 hours is often suitable).
Background reduction: Include a peroxidase blocking step and ensure adequate blocking with appropriate serum (5-10% normal serum from the species of the secondary antibody).
Counterstaining: Use light hematoxylin counterstaining to avoid obscuring specific staining of RNF103.
Rigorous validation of RNF103 antibody specificity should include multiple approaches:
Genetic validation: Use RNF103 knockout or knockdown systems (CRISPR/Cas9, siRNA) to confirm loss of signal.
Multiple antibody approach: Verify results using different antibodies targeting distinct epitopes of RNF103.
Immunogen blocking: Pre-incubate the antibody with excess immunizing peptide to demonstrate competitive inhibition of specific binding.
Mass spectrometry correlation: For pull-down or immunoprecipitation applications, confirm target identity by mass spectrometry.
Signal localization: Confirm that subcellular localization matches expected distribution (endoplasmic reticulum membrane for RNF103) .
Molecular weight verification: Confirm that the detected band corresponds to the expected molecular weight of RNF103 or its known isoforms.
Cross-species validation: If the antibody is reported to react with multiple species, confirm consistent results across those species while accounting for potential species-specific differences in expression patterns.
When encountering non-specific binding with RNF103 antibodies, researchers can implement several strategies:
Titrate the antibody concentration: Testing several dilutions beyond the recommended range can often identify an optimal concentration that maximizes specific binding while minimizing background.
Modify blocking conditions: Try alternative blocking agents (BSA, normal serum, commercial blockers) or increase blocking time/concentration.
Adjust buffer composition: Adding non-ionic detergents (0.1-0.3% Triton X-100), increasing salt concentration (150-500 mM NaCl), or adding competing proteins can reduce non-specific interactions.
Pre-adsorption: For tissues with high endogenous biotin or cross-reactivity concerns, pre-adsorb the antibody with tissue powder from the experimental species.
Alternative secondary antibody: Try secondary antibodies from different vendors or with different conjugates to address potential cross-reactivity issues.
Reduce exposure time: For detection methods with variable exposure (chemiluminescence, fluorescence), optimize exposure time to capture specific signal before background becomes problematic.
Apply computational approaches: For particularly challenging samples, consider using computer-assisted analysis to separate signal from background based on intensity profiles and localization patterns .
Investigating RNF103's function in the ERAD pathway requires multi-faceted experimental approaches:
Protein-protein interaction studies:
Co-immunoprecipitation using RNF103 antibodies to identify binding partners
Proximity labeling methods (BioID, APEX) with RNF103 as the bait protein
Split-ubiquitin yeast two-hybrid assays suitable for membrane proteins
Ubiquitination assays:
In vitro ubiquitination assays with purified components
Cell-based ubiquitination assays with His-tagged ubiquitin pulldowns
Chain-specific ubiquitin antibodies to determine ubiquitin chain topology on substrates
Functional studies:
RNF103 depletion or overexpression followed by proteasome inhibition to identify accumulated substrates
Pulse-chase experiments to measure protein degradation rates in the presence/absence of RNF103
ER stress response monitoring using reporters (e.g., XBP1 splicing, CHOP induction)
Structural studies:
Domain mapping using truncation mutants and RNF103 antibodies
Point mutations in the RING domain to disrupt E3 ligase activity
Subcellular localization studies to confirm ER membrane positioning
Disease models:
Combining RNF103 antibodies with advanced biophysical methods can provide deeper insights into protein function:
Förster Resonance Energy Transfer (FRET): Using fluorescently labeled RNF103 antibodies (or their fragments) paired with fluorescently labeled potential interaction partners to detect proximity in living cells.
Biolayer Interferometry (BLI) or Surface Plasmon Resonance (SPR): Immobilizing RNF103 antibodies to capture the protein from lysates, then measuring binding kinetics with potential substrate proteins or E2 ubiquitin-conjugating enzymes.
Single-molecule imaging: Using quantum dot-conjugated antibodies against RNF103 to track its dynamics and interactions in live cells at the single-molecule level.
Mass spectrometry coupled approaches:
Proximity-dependent biotin identification (BioID) followed by streptavidin pulldown and mass spectrometry
Crosslinking mass spectrometry (XL-MS) with RNF103 antibodies for immunoprecipitation
Hydrogen-deuterium exchange mass spectrometry (HDX-MS) to identify regions involved in protein-protein interactions
Cryo-electron microscopy: Using RNF103 antibodies for immunogold labeling to identify the protein within larger complexes visualized by cryo-EM.
Antibody-based proximity proteomics: Techniques like selective proteomic proximity labeling using tyramide (SPPLAT) that combine antibody specificity with radical-based labeling of proximal proteins .
Modern computational methods offer significant advantages for RNF103 antibody research:
Epitope prediction and antibody design: Computational algorithms can predict immunogenic epitopes of RNF103 and guide the design of more specific antibodies with reduced cross-reactivity .
Specificity profile customization: Computational modeling can help develop antibodies with either high specificity for RNF103 alone or controlled cross-specificity for multiple target ligands, as demonstrated in recent research .
Binding mode identification: Machine learning approaches can identify different binding modes associated with particular ligands, allowing researchers to disentangle these modes even when they involve chemically similar epitopes .
Library optimization: Computational analysis of phage display data can guide the creation of antibody libraries with improved coverage of potential binding specificities .
Cross-reactivity prediction: In silico methods can predict potential cross-reactivities with other proteins containing similar structural motifs to the RNF103 RING-H2 finger domain.
Structure-guided antibody engineering: Using predicted or experimentally determined structures of RNF103 to design antibodies that target functional domains with improved specificity.
Multiple bands in Western blot using RNF103 antibodies may have several explanations:
Alternative splicing: RNF103 undergoes alternative splicing resulting in multiple transcript variants , potentially producing protein isoforms of different molecular weights.
Post-translational modifications: Ubiquitination, phosphorylation, or glycosylation can alter the apparent molecular weight of RNF103.
Proteolytic processing: Partial degradation during sample preparation may generate fragments recognized by the antibody.
Cross-reactivity: The antibody may recognize related proteins with similar epitopes, particularly other RING finger proteins.
Incomplete denaturation: Membrane proteins like RNF103 can form aggregates if not fully denatured, appearing as higher molecular weight bands.
To address this issue:
Compare observed band patterns with known isoforms and modifications of RNF103
Optimize sample preparation to minimize protein degradation
Test different denaturing conditions (temperature, detergents, reducing agents)
Consider using more specific antibodies targeting unique regions of RNF103
Validate with knockout/knockdown controls to identify which bands represent specific detection
Detecting low-abundance RNF103 in tissues requires sensitivity optimization:
Sample enrichment:
Signal amplification methods:
Tyramide signal amplification (TSA) for immunohistochemistry
Enhanced chemiluminescence (ECL) substrates with higher sensitivity for Western blot
Quantum dot-conjugated secondary antibodies for fluorescence detection
Reduced background strategies:
Extended blocking times (overnight at 4°C)
Higher BSA concentrations in wash and antibody dilution buffers (3-5%)
Addition of non-specific proteins from the host species of the secondary antibody
Protocol modifications:
Extended primary antibody incubation (48-72 hours at 4°C)
Optimized antigen retrieval methods for fixed tissues
Signal converter enzymes for chromogenic detection
Alternative detection methods:
Proximity ligation assay (PLA) to visualize protein interactions with higher sensitivity
RNAscope combined with immunofluorescence to correlate mRNA and protein expression
Tissue-specific optimization for RNF103 antibody applications:
| Tissue Type | Common Challenges | Optimization Strategies |
|---|---|---|
| Brain (high RNF103 expression) | High lipid content affecting fixation | Use shorter fixation times; perform antigen retrieval with Tris-EDTA buffer pH 9.0 |
| Peripheral tissues (lower expression) | Weak signal | Implement signal amplification; increase antibody concentration; extend incubation time |
| Highly vascular tissues | High background due to endogenous peroxidases | Double peroxidase quenching step; use fluorescent detection instead of HRP |
| Adipose tissue | Non-specific binding, high autofluorescence | Use sudan black to reduce autofluorescence; extend blocking time |
| Muscle tissue | High background | Add avidin/biotin blocking step; use higher salt concentration in wash buffers |
| Embryonic/developmental tissues | Different expression patterns | Optimize fixation for developmental stage; compare with in situ hybridization |
Additionally, tissue-specific fixation protocols may need adjustment, as overfixation can mask epitopes, particularly in dense tissues, while underfixation in delicate tissues can lead to poor morphology.
When selecting an RNF103 antibody for specific applications, researchers should consider:
RNF103 antibodies are helping elucidate potential connections between ER stress, protein degradation, and neurodegenerative conditions:
Depression and antidepressant mechanisms: Given the induction of RNF103 expression by electroconvulsive therapy and antidepressant treatments , antibodies enable tracking of protein expression changes in relevant brain regions, potentially revealing new therapeutic targets.
ER stress in neurodegeneration: As an ERAD component, RNF103 may play a role in managing misfolded proteins implicated in conditions like Alzheimer's and Parkinson's diseases. Antibodies allow researchers to:
Quantify RNF103 expression changes during disease progression
Identify co-localization with disease-associated misfolded proteins
Track changes in RNF103 distribution in affected neurons
Protein quality control pathways: Dysfunction in protein degradation pathways is implicated in multiple neurodegenerative conditions. RNF103 antibodies help map the protein's interactions with:
ER chaperones that recognize misfolded proteins
Other ERAD components in the degradation machinery
Substrates that accumulate when the system is compromised
Therapeutic target assessment: As potential therapeutic strategies targeting the ubiquitin-proteasome system emerge, RNF103 antibodies provide essential tools for:
Validating target engagement in drug development
Monitoring on-target and off-target effects of candidate compounds
Establishing biomarkers for treatment response
Several cutting-edge technologies are advancing RNF103 antibody development:
Phage display with high-throughput sequencing: This approach allows systematic identification of antibodies with customized specificity profiles, enabling the design of antibodies with either high specificity for RNF103 or controlled cross-reactivity with related proteins .
Computational antibody design: Biophysics-informed modeling combined with machine learning algorithms can predict binding modes and specificity profiles, allowing in silico screening and optimization before experimental validation .
Nanobody and single-domain antibody technology: These smaller antibody formats may offer improved access to constrained epitopes within membrane proteins like RNF103, potentially enhancing detection of the native conformation.
Site-specific conjugation methods: Advanced chemical biology techniques allow precise conjugation of detection molecules to specific sites on antibodies, improving orientation and reducing functional interference.
Multiparametric antibody engineering: Designing antibodies with conditional binding properties that respond to pH, redox state, or the presence of specific cofactors could enable more sophisticated experimental applications.
Spatially-resolved antibody-based proteomics: Combining RNF103 antibodies with spatial transcriptomics or imaging mass cytometry can provide unprecedented insights into protein localization and interaction networks in complex tissues.
Integrating RNF103 antibodies into multi-omics frameworks enables comprehensive understanding of its biological roles:
Proteogenomic integration:
Correlating RNF103 protein levels (detected by antibodies) with mRNA expression data
Mapping post-translational modifications of RNF103 and correlating with regulatory mechanisms
Identifying discrepancies between transcriptome and proteome data that might indicate specialized regulation
Structural biology connections:
Using antibodies to stabilize specific conformations of RNF103 for structural studies
Validating in silico structural predictions with epitope accessibility studies
Mapping functional domains through selective antibody binding
Interactome analysis:
Antibody-based pull-downs coupled with mass spectrometry to identify interaction partners
Validation of high-throughput interaction data from yeast two-hybrid or proximity labeling studies
Temporal analysis of dynamic protein complexes under different cellular conditions
Single-cell multi-omics:
Combining antibody-based protein detection with single-cell RNA sequencing
Correlating RNF103 expression with cellular phenotypes at single-cell resolution
Identifying cell type-specific functions and regulatory networks
Systems biology approaches:
Positioning RNF103 within signaling networks through antibody-based quantification after perturbations
Comparing experimental antibody-based data with computational predictions of network behavior
Developing predictive models of RNF103 function in cellular stress responses
Thorough reporting of RNF103 antibody details enhances reproducibility:
Antibody identification:
Commercial source, catalog number, and lot number
For custom antibodies: immunogen sequence, host species, and production method
RRID (Research Resource Identifier) when available
Validation methods:
Describe specificity controls used (knockout/knockdown validation, immunogen blocking)
Reference previous publications demonstrating validity for the specific application
Include representative images of validation experiments in supplementary materials
Experimental conditions:
Complete protocol details including fixation, blocking, dilutions, and incubation conditions
Buffer compositions and any critical reagents
Detection methods and image acquisition parameters
Quantification methods:
Software and algorithms used for image analysis
Statistical approaches for interpreting antibody-based quantitative data
Normalization procedures and controls
Limitations and potential artifacts:
Known cross-reactivities or limitations of the antibody
Potential confounding factors in the experimental system
Alternative interpretations of the results
The research horizon for RNF103 antibodies includes several promising directions:
Therapeutic antibody development: Engineering antibodies that can modulate RNF103 activity could have therapeutic potential in conditions where ER stress and protein quality control are dysregulated.
Single-molecule dynamics: Using antibody fragments to track RNF103 movement and interactions in living cells at nanoscale resolution.
Substrate identification: Developing antibody-based proximity labeling techniques specific for RNF103 to identify its ubiquitination substrates in different tissues and under various stress conditions.
Conditional knockdowns: Antibody-based targeted protein degradation approaches (e.g., TRIM-Away) to achieve acute depletion of RNF103 in specific cellular compartments.
Biomarker development: Exploring whether RNF103 levels or modifications could serve as biomarkers for ER stress-related diseases or treatment responses.
Engineered biosensors: Creating antibody-based fluorescent biosensors that report on RNF103 conformation or activity changes in real-time.
Cross-species comparative studies: Using highly specific antibodies to compare RNF103 functions across evolutionary diverse organisms to understand conserved and specialized roles.