The designation "AUR3" may stem from typographical errors or misinterpretations of established Aurora kinase nomenclature:
Aurora A (AURKA): Serine/threonine kinase regulating mitosis; overexpressed in cancers like breast and colorectal .
Aurora B (AURKB/AIM1): Chromosomal passenger protein critical for cytokinesis .
Aurora C (AURKC): Primarily involved in meiosis; limited somatic tissue expression.
No Aurora kinase isoform is classified as "AUR3" in the HUGO Gene Nomenclature Committee (HGNC) or UniProt databases .
Immunogen: Recombinant human AURKA fragments (e.g., Met1-Glu131) or synthetic phosphopeptides (e.g., Thr288) .
Specificity:
Functional Roles:
#28711: Validated for detecting endogenous Aurora B (~40 kDa) in human cells .
Mechanism: Targets the amino-terminal region to study mitotic regulation .
Orai3 Antibody (ab254260): Targets a calcium channel protein unrelated to Aurora kinases .
Aureolysin (aur) Antibody: Binds Staphylococcus aureus metallopeptidase, not Aurora kinases .
Key parameters for Aurora antibody validation include:
AUR3 Antibody is a monoclonal antibody developed against human Aurora-A serine/threonine kinase. This antibody binds specifically to centrosomes and spindle poles in human cells, making it valuable for studying cell division processes. When properly validated, the antibody should detect a single protein band at approximately 46 kDa on SDS-PAGE Western blots . Aurora kinase antibodies are critical for investigating cell cycle regulation, particularly mitotic events involving centrosome duplication and spindle formation.
When selecting an AUR3 Antibody, researchers should evaluate its validation data across multiple applications. Effective antibodies should: (1) recognize the target protein in Western blots at the correct molecular weight, (2) show specific localization to centrosomes and spindle poles in immunofluorescence, (3) successfully immunoprecipitate the kinase, and (4) ideally cross-react with orthologs from model organisms if comparative studies are planned . Additionally, researchers should consider whether the antibody affects kinase activity, as some antibodies may inhibit function while others (like the 35C1 described in literature) allow measurement of kinase activity after immunoprecipitation .
A robust validation protocol for AUR3 Antibody should include:
| Validation Method | Procedure | Expected Result |
|---|---|---|
| ELISA | Test against purified recombinant His-tagged Aurora-A | Positive binding signal |
| Western Blot | Test against recombinant protein and cell lysates | Single band at 46 kDa |
| Immunofluorescence | Staining of cultured cells | Specific binding to centrosomes and spindle poles |
| Knockout Controls | Testing in Aurora-A knockout cells | Absence of signal |
| Cross-species reactivity | Testing against mouse Aurora-A | Positive reaction if conserved epitope |
The most definitive validation involves using genetic knockout models where Aurora-A is absent, confirming that the antibody signal disappears in these samples . Organizations like YCharOS are working to characterize antibodies using knockout cell lines, providing independent validation data that researchers should consult when available .
For optimal immunofluorescence using AUR3 Antibody:
Culture cells on appropriate coverslips or chamber slides
Fix cells using 4% paraformaldehyde (10 minutes at room temperature) or ice-cold methanol (5 minutes)
Permeabilize with 0.1% Triton X-100 (if using paraformaldehyde fixation)
Block with 5% normal serum in PBS for 1 hour
Incubate with optimally titrated AUR3 Antibody (typically 1:100 to 1:500 dilution) overnight at 4°C
Wash three times with PBS
Apply fluorophore-conjugated secondary antibody at appropriate dilution
Counterstain DNA with DAPI
Mount and image using confocal microscopy
When selecting secondary antibodies and fluorophores, consider the expression level of Aurora-A (which varies through the cell cycle) and match with appropriately bright fluorophores for clear visualization of centrosomes . For co-staining experiments, avoid fluorophores with high spectral overlap when detecting markers that might co-localize with Aurora-A .
For Western blotting, researchers should:
Harvest cells during mitosis when Aurora-A expression peaks
Use phosphatase inhibitors in lysis buffers to preserve phosphorylation states
Titrate the antibody to determine optimal concentration (typically 1:1000 to 1:5000)
Include positive controls (recombinant Aurora-A) and negative controls (Aurora-A depleted samples)
Consider using gradient gels (4-12%) for optimal separation
Block membranes with 5% BSA rather than milk (which contains phosphatases)
Incubate with primary antibody overnight at 4°C
Use enhanced chemiluminescence detection systems for highest sensitivity
To prevent non-specific binding, centrifuge the antibody at 10,000 RPM for 3 minutes prior to dilution, particularly important if using conjugated antibodies that can form aggregates .
Every experiment with AUR3 Antibody should include:
| Control Type | Purpose | Implementation |
|---|---|---|
| Positive Control | Confirms antibody functionality | Include recombinant Aurora-A protein or cell line with known Aurora-A expression |
| Negative Control | Assesses non-specific binding | Include Aurora-A knockout/knockdown cells or isotype control antibody |
| Secondary-only Control | Evaluates background from secondary antibody | Omit primary antibody |
| Titration Control | Optimizes signal-to-noise ratio | Test several dilutions to find optimal concentration |
| Absorption Control | Confirms epitope specificity | Pre-incubate antibody with excess target peptide before staining |
The absence of these controls significantly increases the risk of misinterpreting experimental results, especially when studying a spatially and temporally regulated protein like Aurora-A .
When incorporating AUR3 Antibody into flow cytometry panels:
For panels with more than 8 markers, consider using spectral flow cytometry platforms like Cytek Aurora, which provides better resolution for discriminating between similar fluorophores .
Detecting phosphorylated Aurora kinase requires special considerations:
Use specialized fixation buffers designed for phosphoprotein preservation
Ensure rapid sample processing to prevent phosphatase activity
Keep samples cold (4°C) during all processing steps
Use phosphatase inhibitors in all buffers
Select antibodies specifically validated for phospho-epitope detection
Include positive controls treated with phosphatase inhibitors
Include negative controls treated with phosphatases or Aurora kinase inhibitors
Consider using dual staining with total Aurora-A and phospho-specific antibodies to normalize for expression variations
Phosphorylation states are particularly sensitive to experimental conditions, making standardized protocols essential for reproducible results across experiments .
For immunoprecipitation (IP) followed by kinase activity assays:
Confirm the antibody does not inhibit kinase activity (as verified for 35C1 clone)
Prepare cell lysates in non-denaturing buffers containing protease and phosphatase inhibitors
Pre-clear lysates with protein A/G beads to reduce non-specific binding
Incubate cleared lysates with optimized amount of AUR3 Antibody (typically 1-5 μg)
Capture antibody-antigen complexes with protein A/G beads
Wash extensively to remove contaminants while preserving kinase activity
Split the immunoprecipitate for parallel analysis:
Western blot to confirm Aurora-A pull-down
Kinase assay using appropriate substrates (e.g., histone H3)
Quantify phosphorylation by autoradiography or phospho-specific antibodies
The 35C1 monoclonal antibody described in the literature specifically maintains Aurora-A kinase activity after immunoprecipitation, making it suitable for measuring in vivo kinase activity .
Machine learning (ML) approaches can enhance AUR3 Antibody experiments in several ways:
Predicting antibody-antigen binding affinities using library-on-library approaches
Optimizing experimental design through active learning strategies, which can reduce the number of required experiments by up to 35%
Improving out-of-distribution prediction for novel Aurora kinase variants or related proteins
Analyzing complex binding patterns in high-dimensional data from flow cytometry or imaging experiments
Developing more specific antibodies by analyzing epitope characteristics that confer specificity
The Absolut! simulation framework, for example, has been used to evaluate active learning strategies for antibody-antigen binding prediction, potentially accelerating experimental workflows . By implementing these computational approaches, researchers can design more efficient experimental protocols and reduce the resources needed for comprehensive analyses.
Researchers should be aware of these common pitfalls:
To avoid these issues, researchers should validate findings with multiple detection methods and include appropriate genetic controls (siRNA knockdown or CRISPR knockout) . Additionally, understanding the normal cell cycle-dependent expression pattern of Aurora-A is crucial for correct interpretation.
When troubleshooting weak or absent signals:
| Problem | Potential Causes | Solutions |
|---|---|---|
| Weak signal | Suboptimal antibody concentration | Perform antibody titration |
| Low target expression | Enrich for mitotic cells or use synchronization | |
| Inefficient epitope access | Try different fixation/permeabilization methods | |
| Antibody degradation | Use fresh aliquots, avoid freeze-thaw cycles | |
| No signal | Wrong application | Verify antibody is validated for your application |
| Incorrect species reactivity | Confirm cross-reactivity with your model system | |
| Epitope masking | Try epitope retrieval methods | |
| Secondary antibody mismatch | Ensure compatible isotype and species |
For flow cytometry applications specifically, fluorochrome aggregation can reduce signal quality. Centrifuge antibody solutions at high speed (10,000 RPM for 3 minutes) before use to remove aggregates, and use specialized staining buffers for antibodies conjugated to polymer dyes like Brilliant Violet .
To ensure batch-to-batch consistency:
Maintain a reference stock of a validated antibody lot
Test each new batch side-by-side with the reference in your specific application
Compare staining patterns, signal intensity, and background levels
Document lot numbers and performance metrics for all experiments
Consider developing a standardized quantitative assay for antibody validation
Store validation data in a centralized database for reference
Participate in community efforts like YCharOS to share antibody validation data
Antibody reproducibility has been identified as a major challenge in biomedical research, with many antibodies failing to recognize their intended targets or recognizing additional molecules . Careful validation of each batch is therefore essential for research integrity.
Cryo-electron microscopy (cryoEM) is revolutionizing antibody research through:
Structure-based antibody discovery approaches that can identify functional antibody sequences
Detailed epitope mapping at near-atomic resolution
Conformational analysis of antibody-antigen complexes
Evaluation of binding modes and interaction surfaces
Optimization of antibody properties based on structural insights
These approaches can potentially improve Aurora kinase antibodies by enabling rational design of reagents with enhanced specificity and reduced cross-reactivity with related family members. CryoEM can also help characterize the structural basis for antibodies that either inhibit or preserve Aurora kinase activity .
Several initiatives are addressing antibody reliability challenges:
YCharOS works with antibody manufacturers and knockout cell line producers to independently validate antibodies, identifying high-performing renewable antibodies for many targets
The "Only Good Antibodies" initiative brings together researchers and partner organizations to promote best practices in antibody selection and validation
Open data sharing platforms for antibody characterization results
Community-driven validation efforts to identify reliable reagents
Development of standardized validation protocols across research communities
These initiatives aim to address technical, policy, behavioral, and data sharing challenges in antibody research. By participating in these collaborative efforts, researchers can contribute to improved research reproducibility and integrity in the field .