ARI2 antibodies are immunoreagents designed to detect and analyze ARIH2, an E3 ubiquitin ligase mediating Lys-48- and Lys-63-linked polyubiquitination, leading to proteasomal degradation of target proteins . ARIH2 is critical for embryogenesis, immune homeostasis, and cancer progression .
ARI2 antibodies have been instrumental in uncovering ARIH2’s functional roles:
Western Blotting: Use 10–20 µg lysate per lane; recommended dilutions 1:1000–1:4000 .
Immunoprecipitation: Optimal results with 1–3 mg total protein lysate .
Validation: Antibodies show high affinity (KD values 1–2 orders better than traditional monoclonals) .
Current studies focus on ARIH2’s role in tumor suppression, viral pathogenesis, and immune modulation. Antibodies like EPR7670 (ab133744) enable high-resolution imaging of ARIH2 in mitotic chromatin and DNA repair complexes .
ARID2 (AT-rich interactive domain 2) is a 197 kDa protein involved in chromatin remodeling complexes. It functions as a subunit of the PBAF (Polybromo-associated BAF) complex, which regulates gene expression through ATP-dependent chromatin remodeling. The protein contains 1835 amino acids and plays critical roles in transcriptional regulation by facilitating accessibility of transcription factors to DNA. When working with ARID2 antibodies, it's important to understand that the observed molecular weight on western blots typically ranges from 200-230 kDa, which is slightly higher than the calculated weight due to post-translational modifications .
ARID2 antibody has been validated for several experimental applications:
Application | Recommended Dilution | Notes |
---|---|---|
Western Blot (WB) | 1:500-1:1000 | Positive signals detected in Jurkat and K-562 cells |
Immunohistochemistry (IHC) | 1:250-1:1000 | Positive detection in mouse testis tissue |
ELISA | Application-dependent | Requires optimization for specific assay conditions |
For optimal results in IHC applications, antigen retrieval with TE buffer at pH 9.0 is recommended, though citrate buffer at pH 6.0 may also be used as an alternative .
ARID2 antibody should be stored at -20°C where it remains stable for one year after shipment. The antibody is typically supplied in PBS with 0.02% sodium azide and 50% glycerol at pH 7.3. Aliquoting is not necessary for -20°C storage. For smaller 20μl sizes that contain 0.1% BSA, avoid repeated freeze-thaw cycles to maintain antibody activity. When handling the antibody, wear appropriate PPE due to the presence of sodium azide, which is toxic .
ARID2 antibody has been tested and validated with:
Sample Type | Validation Status |
---|---|
Human cell lines | Positive: Jurkat cells, K-562 cells |
Mouse tissues | Positive: Mouse testis tissue |
Other species | Limited data available |
When extending to untested samples, preliminary validation experiments are strongly recommended to confirm reactivity and specificity .
Epitope mapping for ARID2 antibody can be approached similarly to methods used for other antibodies, such as those described for ACE2 autoantibodies. A comprehensive approach would involve:
Creating a peptide library spanning the entire ARID2 protein (1835 amino acids)
Synthesizing overlapping peptides (e.g., 15 amino acids with 11 amino acid overlaps)
Immobilizing peptides on glass microarray surfaces using hydrophilic linker moieties
Incubating diluted antibody samples (typically 1:200) with the peptide microarray
Detecting binding with fluorescently labeled secondary antibodies
Analyzing binding patterns to identify specific epitopes
This approach can reveal which domains of ARID2 are recognized by the antibody, which is particularly valuable for determining if functional domains are targeted and if the antibody might interfere with protein-protein interactions .
When encountering cross-reactivity with ARID2 antibody, consider implementing these advanced troubleshooting strategies:
Perform pre-adsorption tests by incubating the antibody with purified recombinant ARID2 protein before application to samples
Increase blocking stringency by using 5% BSA or specialized blocking reagents designed for polyclonal antibodies
Optimize antibody concentration through careful titration experiments
Include additional washing steps with higher salt concentrations (up to 500 mM NaCl) to reduce non-specific binding
Validate results with multiple antibodies targeting different epitopes of ARID2
Consider using knockout or knockdown controls to definitively establish specificity
Cross-reactivity assessment is particularly important when studying closely related ARID family proteins that share sequence homology with ARID2 .
Machine learning models can significantly enhance ARID2 antibody applications through improved prediction of binding characteristics:
Library-on-library approaches can identify specific interacting pairs between ARID2 antibody variants and target epitopes
Active learning algorithms can reduce experimental costs by starting with a small labeled subset of binding data and iteratively expanding only the most informative data points
Out-of-distribution prediction can help estimate how ARID2 antibodies might interact with previously uncharacterized mutant proteins or related family members
Simulation frameworks like Absolut! can be used to evaluate potential binding before committing to expensive experimental validation
Studies have shown that optimized active learning strategies can reduce the number of required antigen mutant variants by up to 35% and accelerate the learning process compared to random sampling approaches .
For multiplex immunofluorescence studies incorporating ARID2 antibody, researchers should address these critical factors:
Antibody Clone Selection:
Verify the rabbit polyclonal ARID2 antibody (23406-1-AP) is compatible with your multiplexing approach
Consider potential cross-reactivity with other antibodies in your panel
Signal Amplification:
For nuclear proteins like ARID2, tyramide signal amplification may be necessary due to lower abundance
Optimize signal-to-noise ratio through careful titration experiments
Antigen Retrieval Compatibility:
Ensure all antibodies in the multiplex panel are compatible with the TE buffer pH 9.0 recommended for ARID2
If incompatibilities exist, sequential staining approaches may be required
Spectral Overlap:
Design panels accounting for the nuclear localization of ARID2 to avoid spatial overlap with other nuclear markers
Perform single-color controls to establish proper unmixing parameters
Validation Controls:
For optimal Western blot detection of ARID2, follow these methodological guidelines:
Sample Preparation:
Use RIPA buffer supplemented with protease inhibitors for efficient extraction
Include phosphatase inhibitors if phosphorylation status is relevant
Heat samples at 95°C for 5 minutes in loading buffer containing SDS and DTT
Gel Selection and Transfer:
Use 6-8% SDS-PAGE gels or gradient gels (4-15%) to resolve the 200-230 kDa ARID2 protein
Perform wet transfer at low voltage (30V) overnight at 4°C to ensure complete transfer of high molecular weight proteins
Use PVDF membrane (0.45 μm pore size) rather than nitrocellulose for better retention of large proteins
Antibody Incubation:
Block membranes with 5% non-fat dry milk in TBST for 1 hour at room temperature
Dilute ARID2 antibody 1:500 in blocking buffer for overnight incubation at 4°C
Wash extensively (4 × 10 minutes) with TBST before secondary antibody incubation
Detection Optimization:
Use enhanced chemiluminescence with extended exposure times (up to 5 minutes)
Consider gradient exposure times to capture the optimal signal intensity
For quantitative analysis, use fluorescently-labeled secondary antibodies and fluorescence imaging
This protocol has been validated using Jurkat and K-562 cell lysates, which serve as positive controls for ARID2 detection .
A robust experimental design using ARID2 antibody should incorporate these essential controls:
Control Type | Purpose | Implementation |
---|---|---|
Positive Control | Verify antibody functionality | Include Jurkat or K-562 cell lysates for WB; mouse testis sections for IHC |
Negative Control | Assess non-specific binding | Include samples with ARID2 knockdown or tissues known to not express ARID2 |
Loading Control | Normalize protein loading | Probe for housekeeping proteins (e.g., GAPDH, β-actin) on the same membrane |
Secondary-only Control | Detect non-specific secondary antibody binding | Omit primary antibody but include all other reagents |
Isotype Control | Evaluate background from primary antibody | Use non-specific rabbit IgG at the same concentration |
Peptide Competition | Confirm epitope specificity | Pre-incubate antibody with excess ARID2 immunogen peptide |
These controls help distinguish true positive signals from artifacts and allow proper interpretation of experimental results when using ARID2 antibody .
Optimizing epitope accessibility for ARID2 IHC requires careful consideration of fixation and antigen retrieval methods:
Fixation Optimization:
Limit fixation time with 10% neutral-buffered formalin to 24 hours to prevent excessive cross-linking
Consider alternative fixatives such as zinc-based fixatives that preserve epitopes while maintaining tissue morphology
For frozen sections, use acetone fixation for 10 minutes at -20°C to improve epitope preservation
Antigen Retrieval Methods:
Heat-induced epitope retrieval (HIER) with TE buffer at pH 9.0 is recommended as the primary method
Alternative protocol: citrate buffer at pH 6.0 with pressure cooking for 15 minutes
Enzymatic retrieval using proteinase K (10 μg/ml for 10-15 minutes) may be tested if HIER is ineffective
Signal Enhancement Strategies:
Implement tyramide signal amplification for low-abundance detection
Extend primary antibody incubation to overnight at 4°C using a 1:250 dilution
Add 0.1% Triton X-100 to antibody diluent to improve tissue penetration
Background Reduction:
Include 10% normal goat serum in blocking buffer
Add 0.1% BSA to washing buffers to reduce non-specific binding
Consider avidin-biotin blocking steps if using biotin-based detection systems
These optimizations have proven effective for detecting ARID2 in mouse testis tissue and can be adapted for other tissue types .
When encountering weak or absent ARID2 signals in Western blots, implement this systematic troubleshooting approach:
Protein Extraction Assessment:
Verify complete lysis using stronger extraction buffers (e.g., urea-containing buffers)
Sonicate samples to improve chromatin-associated protein extraction
Confirm protein concentration using Bradford or BCA assays
Transfer Efficiency Analysis:
Verify transfer of high molecular weight proteins using reversible stains (Ponceau S)
Reduce transfer time or voltage if protein is passing through the membrane
Consider semi-dry transfer systems optimized for high molecular weight proteins
Antibody Optimization:
Increase primary antibody concentration (up to 1:250 dilution)
Extend primary antibody incubation to 48 hours at 4°C
Test alternative lots of the same antibody catalog number
Sample-Specific Considerations:
Confirm ARID2 expression in your specific cell type or tissue through transcript analysis
Check literature for expected ARID2 expression levels in your experimental system
Consider that post-translational modifications may alter epitope accessibility
Detection Enhancement:
Use high-sensitivity ECL substrates designed for low-abundance proteins
Increase exposure time during imaging (up to 30 minutes)
Consider using cooled CCD camera systems with integration capabilities
This approach has helped researchers successfully detect the 200-230 kDa ARID2 protein even in challenging sample types .
Non-specific binding is a common challenge when working with ARID2 antibody. Here are typical sources and mitigation strategies:
Source of Non-Specificity | Mitigation Strategy | Implementation Details |
---|---|---|
Insufficient blocking | Optimize blocking conditions | Use 5% BSA instead of milk; extend blocking to 2 hours at room temperature |
Cross-reactivity with related proteins | Increase washing stringency | Add 0.1% SDS to wash buffer; perform 5 × 10 minute washes |
Secondary antibody background | Improve secondary antibody specificity | Use secondary antibodies cross-adsorbed against multiple species |
Protein overloading | Optimize protein concentration | Reduce loading to 20-30 μg per lane; perform loading titration |
Hydrophobic interactions | Modify buffer composition | Add 0.1% Tween-20 to antibody diluent to reduce non-specific hydrophobic binding |
Endogenous biotin (in IHC) | Block endogenous biotin | Apply avidin/biotin blocking kit before primary antibody incubation |
Endogenous peroxidase activity | Quench endogenous enzymes | Incubate samples with 3% H₂O₂ for 10 minutes before antibody addition |
These strategies have been effective in reducing background and improving signal-to-noise ratio in ARID2 detection experiments .
The calculated molecular weight of ARID2 is 197 kDa, but the observed molecular weight on Western blots is typically 200-230 kDa. This discrepancy requires careful interpretation:
Post-Translational Modifications:
Phosphorylation at multiple sites can significantly increase apparent molecular weight
Other modifications (glycosylation, SUMOylation) may contribute to shifts
Different cell types may exhibit varying modification patterns
Technical Considerations:
Use molecular weight markers that extend beyond 200 kDa for accurate size determination
Run gels at lower voltage (80V) for extended time to improve resolution of high molecular weight proteins
Consider using gradient gels (4-15%) for better size separation in the high molecular weight range
Validation Approaches:
Verify specificity through siRNA knockdown of ARID2, which should reduce the intensity of the true ARID2 band
Test antibody on samples from multiple tissues to establish a consistent molecular weight pattern
Compare results with alternative ARID2 antibodies targeting different epitopes
Data Interpretation Guidelines:
Accept bands within the 200-230 kDa range as potentially legitimate ARID2 signal
Be cautious of bands below 190 kDa, which likely represent degradation products or non-specific binding
Document exact conditions when molecular weight varies to identify potential regulatory mechanisms
Understanding these molecular weight variations is crucial for proper data interpretation and can provide insights into the post-translational regulation of ARID2 .
Machine learning techniques offer powerful tools for analyzing complex ARID2 antibody binding data:
Binding Affinity Prediction:
Train models using library-on-library screening data to predict binding between antibody variants and target epitopes
Implement active learning strategies that iteratively identify the most informative experiments to perform
These approaches can reduce experimental costs by up to 35% compared to random sampling
Epitope Mapping Analysis:
Apply clustering algorithms to identify patterns in peptide array binding data
Use feature importance analysis to identify key amino acid residues critical for antibody recognition
Implement transfer learning from similar antibody-antigen systems to improve prediction accuracy
Cross-Reactivity Assessment:
Develop models that predict potential cross-reactivity with related proteins based on sequence similarity
Use out-of-distribution prediction techniques to estimate binding to novel mutant variants
Validate computational predictions with targeted experimental verification
Experimental Design Optimization:
Implement Bayesian optimization to determine optimal antibody concentrations and incubation conditions
Use simulation frameworks to predict experimental outcomes before committing resources
Accelerate the learning process by approximately 28 steps compared to random experimental design
These machine learning approaches can significantly enhance research efficiency and improve the reliability of ARID2 antibody applications in complex experimental systems .
ARID2 antibody can be effectively employed in ChIP studies to investigate chromatin remodeling complexes:
ChIP Protocol Optimization:
Crosslink cells with 1% formaldehyde for 10 minutes at room temperature
Sonicate chromatin to fragments of 200-500 bp
Use 5 μg of ARID2 antibody per ChIP reaction
Incubate antibody-chromatin mixture overnight at 4°C with rotation
Include appropriate controls (IgG, input, positive control regions)
Technical Considerations:
Pre-clear chromatin with protein A/G beads before antibody addition
Use sonication conditions optimized for nuclear proteins
Include detergents (0.1% SDS, 1% Triton X-100) in wash buffers
Verify fragment size distribution by agarose gel electrophoresis
Downstream Applications:
Perform ChIP-seq to map genome-wide ARID2 binding sites
Use ChIP-qPCR to validate binding at specific genomic loci
Combine with RNA-seq to correlate binding with transcriptional outcomes
Data Analysis Approach:
Use peak calling algorithms optimized for transcription factors
Perform motif analysis to identify DNA sequences associated with ARID2 binding
Integrate with public ChIP-seq datasets for other PBAF complex components
This methodology enables researchers to investigate ARID2's role in chromatin remodeling and transcriptional regulation at a genome-wide scale.
For investigating ARID2 protein interactions, consider these methodological aspects:
Co-Immunoprecipitation (Co-IP) Optimization:
Use gentle lysis buffers (e.g., 150 mM NaCl, 50 mM Tris pH 7.5, 0.5% NP-40) to preserve protein complexes
Pre-clear lysates thoroughly to reduce non-specific binding
Incubate 2-5 μg of ARID2 antibody with 500-1000 μg of protein lysate
Include appropriate controls (IgG pulldown, input, reverse Co-IP)
Cross-Linking Strategies:
Consider reversible cross-linkers (DSP, DTBP) to stabilize transient interactions
Optimize cross-linker concentration and incubation time
Include controls to verify cross-linking efficiency
Detection Methods:
Use reciprocal Co-IP to confirm interactions
Perform Western blot analysis with antibodies against known PBAF complex components
Consider mass spectrometry for unbiased identification of interaction partners
Functional Validation:
Verify biological relevance through knockout/knockdown studies
Map interaction domains using truncation mutants
Assess interaction dynamics in response to cellular stimuli
These approaches enable comprehensive characterization of ARID2's role within the PBAF complex and identification of novel interaction partners.
To investigate post-translational modifications (PTMs) of ARID2:
Sample Preparation:
Include phosphatase inhibitors (sodium orthovanadate, sodium fluoride) to preserve phosphorylation
Add deubiquitinase inhibitors (N-ethylmaleimide) for ubiquitination studies
Use HDAC inhibitors (sodium butyrate, trichostatin A) for acetylation analysis
Enrichment Strategies:
Perform immunoprecipitation with ARID2 antibody followed by Western blot with PTM-specific antibodies
Consider phospho-enrichment using TiO₂ or IMAC prior to analysis
Use ubiquitin-specific pulldown (TUBE technology) for ubiquitination studies
Detection Methods:
Employ PTM-specific antibodies (phospho, acetyl, ubiquitin, SUMO) in Western blots
Consider mass spectrometry for comprehensive PTM mapping
Use Phos-tag gels to separate phosphorylated from non-phosphorylated forms
Functional Validation:
Correlate PTM status with ARID2 activity and localization
Generate site-specific mutants to determine PTM function
Investigate enzymes responsible for adding/removing specific PTMs
This methodological approach provides insights into how PTMs regulate ARID2 function within chromatin remodeling complexes.
Several cutting-edge technologies are expanding the utility of ARID2 antibody in advanced research applications:
Proximity Labeling:
BioID or APEX2 fusions to study the ARID2 interactome in living cells
TurboID for rapid labeling of transient interaction partners
Split-BioID for detecting specific protein-protein interactions
Super-Resolution Microscopy:
STORM/PALM techniques to visualize ARID2 localization at nanometer resolution
Expansion microscopy to physically enlarge samples for improved visualization
Lattice light-sheet microscopy for dynamic imaging of ARID2 in living cells
Single-Cell Applications:
CUT&Tag for single-cell profiling of ARID2 chromatin binding
scRNA-seq combined with ARID2 perturbation for functional genomic analysis
Spatial transcriptomics to correlate ARID2 binding with gene expression in tissue context
Artificial Intelligence:
Deep learning for improved image analysis in ARID2 immunofluorescence
Active learning strategies to optimize experimental design
Predictive modeling for antibody-antigen interactions
These technologies represent the frontier of ARID2 research and offer unprecedented insights into its biological functions and regulatory mechanisms .
A comprehensive validation strategy for ARID2 antibody across experimental systems includes:
Genetic Validation:
CRISPR/Cas9 knockout of ARID2 as negative control
siRNA knockdown to demonstrate signal reduction
Rescue experiments with exogenous ARID2 expression
Multi-Antibody Approach:
Compare results using antibodies targeting different ARID2 epitopes
Correlate findings across multiple commercial antibodies
Use monoclonal and polyclonal antibodies to verify observations
Cross-Species Validation:
Test reactivity in human, mouse, and other model organisms
Compare staining patterns across evolutionarily related species
Assess conservation of molecular weight and subcellular localization
Multi-Technique Confirmation:
Correlate Western blot findings with immunohistochemistry results
Verify protein expression with transcript levels (RT-qPCR, RNA-seq)
Combine immunofluorescence with in situ hybridization
This rigorous validation approach ensures reliable and reproducible results across diverse experimental contexts and biological systems.