The AIP/ARA9 antibody targets the aryl hydrocarbon receptor-interacting protein (AIP/ARA9), a 37 kDa immunophilin protein involved in regulating the aryl hydrocarbon receptor (AhR) pathway. This pathway mediates cellular responses to environmental toxins and plays roles in xenobiotic metabolism, immune regulation, and tumor suppression .
| Characteristic | Detail |
|---|---|
| Target Antigen | AIP/ARA9 (FKBP homology domain) |
| Molecular Weight | 37 kDa (observed: 44 kDa in WB) |
| Host Species | Mouse monoclonal |
| Clone Designation | 35-2 |
| Applications | WB, IHC-P, ChIP, Flow Cytometry |
| Validation | 4 species, 9 applications |
The antibody recognizes the FKBP-like domain of AIP/ARA9, critical for its interaction with AhR and HSP90 .
Western Blot (WB): Detects AIP/ARA9 at 1:1000 dilution in HEK293 lysates .
Immunohistochemistry (IHC-P): Stains paraffin-embedded tissues at 1:500–1:1000 dilution, distinguishing low vs. high AIP expression in pituitary samples .
Flow Cytometry: Requires 1 µg per million cells for surface staining .
Chromatin Immunoprecipitation (ChIP): Effective at 1:10–1:500 dilution for studying DNA-protein interactions .
Specificity: No cross-reactivity with unrelated proteins confirmed via Simple Western and HEK293T/17 cell lysate testing .
Reproducibility: Validated across 33 peer-reviewed studies, including investigations into pituitary adenomas and toxin response pathways .
Pituitary Adenomas: High AIP/ARA9 expression correlates with aggressive tumor behavior, making this antibody a diagnostic tool for identifying high-risk subtypes .
Toxin Response: AIP/ARA9 knockdown studies using this antibody revealed impaired AhR nuclear translocation, confirming its role in xenobiotic detoxification .
| Application | Dilution Range | Notes |
|---|---|---|
| Western Blot | 1:1000 | Band observed at 37–44 kDa |
| IHC-Paraffin | 1:500–1:1000 | Optimized for human pituitary tissue |
| Flow Cytometry | 1 µg/1M cells | Compatible with intracellular staining |
ARI9 Antibody represents a specialized immunological reagent that requires careful validation for specific research applications. When determining appropriate applications, researchers should implement a systematic characterization process including Western blot analysis, immunoprecipitation, immunohistochemistry, and flow cytometry. For optimal results, initial validation should include antibody titration across multiple dilutions (typically 1:100 to 1:10,000) against known positive and negative controls to establish specificity parameters. Cross-reactivity assessment should be performed using related protein targets to ensure binding specificity before proceeding to experimental applications. Additionally, researchers should verify epitope conservation across species if cross-species reactivity is relevant to experimental design.
Determining optimal working concentrations for ARI9 Antibody requires systematic titration experiments across multiple applications. Researchers should begin with a broad concentration range (typically 0.1-10 μg/mL for Western blots, 1-10 μg/mL for immunohistochemistry, and 0.5-5 μg/mL for flow cytometry) and progressively narrow to identify the concentration that maximizes signal-to-noise ratio . Critical to this process is the inclusion of appropriate positive and negative controls to distinguish specific binding from background. For microarray applications, researchers should test antibody concentrations between 1.5-5.0 mg/mL, as this range has been shown to significantly impact printing quality and assay performance . Experimental designs should incorporate factorial approaches testing concentration alongside other variables like buffer composition and incubation conditions to determine optimal working parameters.
Maintaining ARI9 Antibody functionality requires strict adherence to proper storage and handling protocols. Antibodies should be stored at -20°C to -80°C for long-term preservation, with working aliquots maintained at 4°C for periods not exceeding two weeks . Multiple freeze-thaw cycles significantly reduce antibody activity; therefore, single-use aliquots are strongly recommended. To enhance stability, consider adding protein stabilizers such as BSA (0.1-1%) and cryoprotectants like glycerol (30-50%) which prevents freezing at -20°C while maintaining antibody structure. Working solutions should contain preservatives such as sodium azide (0.02-0.05%) to prevent microbial contamination. Researchers should regularly monitor antibody performance for signs of degradation, including decreased signal intensity, increased background, or precipitation, which necessitate replacement with fresh aliquots.
Antibody persistence varies significantly depending on the target antigen, antibody class, and host factors, with important implications for longitudinal research designs. Viral-specific antibodies demonstrate markedly different persistence patterns; for instance, JC virus antibodies typically remain detectable indefinitely, while SARS-CoV-2 antibodies may show substantial decline over time . This variability stems from differences in immune memory establishment, ongoing antigenic stimulation, and antibody stability. A study investigating SARS-CoV-2 antibodies found that individuals with natural infection plus vaccination developed higher anti-RBD antibody levels compared to those with only vaccination or infection alone, demonstrating how exposure patterns influence antibody persistence . When designing experiments with ARI9 Antibody, researchers should consider potential variations in detection sensitivity over time and incorporate appropriate longitudinal sampling strategies to account for possible fluctuations in antibody levels, particularly in monitoring studies.
The three-dimensional structure of antibodies critically influences their stability and functionality, with particular importance attached to complementarity-determining regions (CDRs). Research in antibody structure prediction has identified the CDR-H3 loop as exhibiting substantial structural variability (RMSD values >2 Å), which significantly impacts binding specificity and affinity . Structural modeling of antibodies frequently encounters challenges including potential modeling artifacts such as cis-amide bonds in CDR loops, D-amino acids, and steric clashes that can distort predictions of antibody-antigen interactions . These structural considerations have direct implications for ARI9 Antibody research, particularly when analyzing epitope binding or engineering for enhanced stability. Researchers should implement computational validation using tools like TopModel to identify potential structural anomalies before proceeding with functional studies . Additionally, biophysical characterization including thermal stability assessment (Tm values), aggregation propensity, and surface charge distribution provides critical information for optimizing experimental conditions.
Optimizing ARI9 Antibody for microarray applications requires careful consideration of multiple technical parameters. Based on experimental studies, antibody concentration significantly impacts microarray quality, with concentrations between 1.5-5.0 mg/mL showing optimal performance depending on the specific application . The printing process represents another critical variable, with parameters such as print-head height (PCV height) directly affecting spot morphology and reproducibility. The table below illustrates a factorial experimental design for optimizing antibody printing parameters:
| Factor Pattern | Antibody Concentration (mg/mL) | PCV Height (cm) |
|---|---|---|
| 33 | 5.0 | 8.57 |
| 11 | 1.5 | -11.43 |
| 31 | 5.0 | -11.43 |
| 22 | 2.5 | 0 |
| 21 | 2.5 | -11.43 |
| 13 | 1.5 | 8.57 |
| 23 | 2.5 | 8.57 |
| 32 | 5.0 | 0 |
| 12 | 1.5 | 0 |
This methodical approach enables researchers to identify optimal printing conditions for minimal lateral and vertical misalignment while maximizing signal intensity and consistency . Researchers should evaluate microarray performance using metrics including spot morphology uniformity, signal-to-noise ratio, and reproducibility (with coefficient of variation <14% considered acceptable for quantitative applications).
Validating ARI9 Antibody specificity requires a multi-faceted approach implementing complementary techniques. Researchers should begin with Western blot analysis using positive control samples containing the target protein alongside negative controls lacking the target. This should be followed by immunoprecipitation coupled with mass spectrometry to confirm target identity . For definitive validation, researchers should employ genetic approaches including CRISPR knockout/knockdown models of the target protein, which provides unambiguous specificity confirmation. Peptide competition assays, where pre-incubation with the target peptide blocks antibody binding, offer additional specificity verification. Cross-reactivity assessment should include testing against closely related proteins with similar sequences. Researchers must document validation results quantitatively rather than relying on subjective assessments, and should maintain validation data for reference throughout the research project to ensure consistent interpretation of experimental results.
Designing multiplexed detection systems with ARI9 Antibody requires careful optimization to prevent cross-reactivity and interference. Based on research findings in antibody microarray development, researchers should first optimize concentration parameters, testing ARI9 Antibody at various concentrations between 1.5-5.0 mg/mL to determine optimal printing conditions . When integrating multiple antibodies into a single detection system, researchers must rigorously evaluate potential cross-reactivity through systematic testing of each antibody against all target antigens. Vertical flow immunoassay systems represent an emerging approach for multiplexed detection, combining features from protein microarrays and paper-based colorimetric systems with sensitivity reaching 1 ng/mL and reproducibility (CV) <14% . For optimal results, include internal calibration standards within each assay run and implement appropriate normalization methods to account for technical variations between detection channels. Statistical tools for cross-reactivity correction should be applied during data analysis to ensure accurate quantification of individual targets within the multiplex system.
Implementing rigorous controls is essential for valid interpretation of ARI9 Antibody experiments. Positive controls should include purified recombinant target protein at known concentrations, cell lines or tissues with verified target expression, and when possible, overexpression systems to evaluate antibody performance at high target concentrations. Negative controls must include samples lacking the target protein, such as knockout cell lines generated through CRISPR-Cas9 technology, which provide definitive confirmation of antibody specificity . Additionally, isotype controls matching the ARI9 Antibody class but lacking target specificity should be employed at identical concentrations to distinguish specific binding from potential Fc-mediated interactions. For immunohistochemistry applications, researchers should include tissue panels with known expression patterns of the target protein. Control experiments should be performed under identical conditions to experimental samples, including identical buffer composition, incubation times, and detection methods to ensure valid comparability between control and experimental results.
Inconsistent results with ARI9 Antibody can stem from multiple sources requiring systematic troubleshooting. Primary factors include antibody degradation due to improper storage, which researchers should address through proper aliquoting and storage at -20°C to -80°C . Lot-to-lot variability represents another significant concern; researchers should record lot numbers and perform validation with each new lot. Sample preparation variability significantly impacts results; standardization of protocols including consistent fixation methods for immunohistochemistry and identical lysis conditions for Western blots is essential. Buffer composition critically affects antibody performance; researchers should evaluate pH (6.0-8.0 range), salt concentration (150-500 mM NaCl), and detergent type/concentration. When troubleshooting, implement a systematic approach testing one variable at a time while maintaining detailed records of all experimental conditions. Additionally, researchers should verify target protein expression levels in their experimental samples, as detection sensitivity varies with target abundance and accessibility.
Accurate determination of ARI9 Antibody binding kinetics and affinity requires specialized biophysical techniques. Surface Plasmon Resonance (SPR) represents the gold standard, allowing real-time, label-free measurement of association (kon) and dissociation (koff) rates by immobilizing the target antigen on a sensor chip and flowing the antibody at multiple concentrations . Bio-Layer Interferometry (BLI) offers an alternative approach with advantages including reduced sample volume requirements and suitability for crude samples. For comprehensive thermodynamic characterization, Isothermal Titration Calorimetry (ITC) provides enthalpy (ΔH) and entropy (ΔS) values in addition to binding affinity. Microscale Thermophoresis (MST) enables affinity measurements with minimal sample amounts and can function with crude lysates, though it requires fluorescent labeling. When implementing these techniques, researchers should use fresh, properly stored antibody preparations and include positive control antibodies with known affinity constants for validation. Experiments should be conducted at physiologically relevant conditions, and data should be evaluated for goodness-of-fit to appropriate binding models.
Predicting ARI9 Antibody structure presents significant challenges that researchers must address through comprehensive computational approaches. Current antibody structure prediction tools including ABlooper, IgFold, DeepAb, and Immunebuilder each have strengths and limitations . Researchers should employ multiple prediction platforms and compare results to identify consistent structural features. Particular attention should focus on the complementarity-determining regions (CDRs), especially CDR-H3, which exhibits substantial conformational variability (RMSD values >2 Å) critical for antigen binding specificity . After generating structural models, researchers must implement rigorous validation to identify potential artifacts including cis-amide bonds in CDR loops, D-amino acids, steric clashes, and bonds with non-physical lengths . The TopModel python package provides valuable tools for identifying such structural anomalies. When analyzing antibody structure, researchers should recognize that a single static structure cannot fully capture the dynamic nature of antibody-antigen interactions, particularly in the highly variable CDR-H3 region, and should consider ensemble approaches that represent multiple conformational states.
Therapeutic antibody trial design offers valuable methodological insights for ARI9 research applications. Clinical antibody studies typically employ a systematic progression from in vitro characterization to in vivo efficacy and safety assessment. For example, recent COVID-19 antibody trials included both treatment studies with infected patients and prophylaxis studies targeting prevention in exposed individuals . The Monoclonal Antibody trial conducted in Tucson employed a single-infusion protocol with 30-day monitoring, providing a model for extended-duration antibody efficacy studies . This approach demonstrated that high-dose specific antibodies offered advantages over convalescent plasma, highlighting the importance of dose optimization in antibody research . For ARI9 Antibody studies, researchers should consider implementing similar methodological approaches, including careful dose-response characterization, extended monitoring periods to assess duration of effect, and comparison with standard reagents to benchmark performance. Additionally, researchers should adopt the practice of clearly defined participant inclusion criteria and outcome measures to enhance reproducibility and interpretability of results.
Emerging technologies offer significant opportunities to advance ARI9 Antibody research. Non-contact printing processes for antibody microarrays enable precise control over spot morphology and density, critical for developing high-sensitivity detection platforms . Vertical flow immunoassay systems combine microarray techniques with paper-based colorimetric detection, achieving sensitivity of 1 ng/mL with reproducibility (CV) <14%, offering potential for rapid, multiplexed ARI9 Antibody applications . Advanced structural analysis technologies including hydrogen-deuterium exchange mass spectrometry and cryo-electron microscopy provide unprecedented insights into antibody-antigen interactions at the molecular level . High-throughput epitope mapping technologies enable comprehensive characterization of binding specificity, critical for understanding ARI9 Antibody function. Computational advances in antibody structure prediction, though still facing challenges with CDR loop modeling, continue to improve in accuracy and reliability . Researchers should consider incorporating these emerging technologies into ARI9 Antibody research projects while maintaining awareness of their current limitations and implementing appropriate validation to ensure reliable results.